(19)
(11)EP 3 283 527 B1

(12)EUROPEAN PATENT SPECIFICATION

(45)Mention of the grant of the patent:
13.01.2021 Bulletin 2021/02

(21)Application number: 16718802.8

(22)Date of filing:  12.04.2016
(51)International Patent Classification (IPC): 
C07K 16/28(2006.01)
A61P 35/00(2006.01)
A61K 39/395(2006.01)
(86)International application number:
PCT/US2016/027038
(87)International publication number:
WO 2016/168149 (20.10.2016 Gazette  2016/42)

(54)

COMBINATION THERAPY FOR CANCER

KOMBINATIONSTHERAPIE GEGEN KREBS

POLYTHÉRAPIE CONTRE LE CANCER


(84)Designated Contracting States:
AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

(30)Priority: 13.04.2015 US 201562146766 P
10.07.2015 US 201562190945 P

(43)Date of publication of application:
21.02.2018 Bulletin 2018/08

(60)Divisional application:
20212116.6

(73)Proprietor: Five Prime Therapeutics, Inc.
South San Francisco, CA 94080 (US)

(72)Inventors:
  • MASTELLER, Emma
    South San Francisco, California 94080 (US)
  • BRENNAN, Thomas
    South San Francisco, California 94080 (US)
  • BELLOVIN, David
    South San Francisco, California 94080 (US)
  • BAKER, Kevin
    South San Francisco, California 94080 (US)
  • WONG, Brian
    South San Francisco, California 94080 (US)

(74)Representative: Mewburn Ellis LLP 
Aurora Building Counterslip
Bristol BS1 6BX
Bristol BS1 6BX (GB)


(56)References cited: : 
WO-A1-2013/132044
WO-A2-2011/140249
WO-A1-2014/036357
WO-A2-2011/140249
  
  • PATEL S ET AL: "Colony-stimulating factor-1 receptor inhibitors for the treatment of cancer and inflammatory disease", CURRENT TOPICS IN MEDICINAL CHEMISTRY, BENTHAM SCIENCE PUBLISHERS LTD.HILVERSUM, NL, vol. 9, no. 7, 1 January 2009 (2009-01-01) , pages 599-610, XP002570335, ISSN: 1568-0266, DOI: 10.2174/156802609789007327
  • WAGNER CARRIE ET AL: "Evaluation of serum biomarkers associated with radiographic progression in methotrexate-naive rheumatoid arthritis patients treated with methotrexate or golimumab", JOURNAL OF RHEUMATOLOGY, JOURNAL OF RHEUMATOLOGY PUBLISHING COMPANY, CA, vol. 40, no. 5, 30 April 2013 (2013-04-30) , pages 590-598, XP009510622, ISSN: 0315-162X, DOI: 10.3899/JRHEUM.120889
  • RICHARDS D M ET AL: "Immune cell activation by novel hexavalent CD40 agonist APG1233 compared to trimeric formats or agonistic anti-CD40 antibodies", EUROPEAN JOURNAL OF CANCER, vol. 69, 29 November 2016 (2016-11-29), XP029843758, ISSN: 0959-8049, DOI: 10.1016/S0959-8049(16)32893-3
  • KNORR DAVID A ET AL: "Toxicity of an Fc-engineered anti-CD40 antibody is abrogated by intratumoral injection and results in durable antitumor immunity.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 23 10 2018, vol. 115, no. 43, 23 October 2018 (2018-10-23), pages 11048-11053, XP002788203, ISSN: 1091-6490
  • Y. ZHU ET AL: "CSF1/CSF1R Blockade Reprograms Tumor-Infiltrating Macrophages and Improves Response to T-cell Checkpoint Immunotherapy in Pancreatic Cancer Models", CANCER RESEARCH, vol. 74, no. 18, 31 July 2014 (2014-07-31) , pages 5057-5069, XP055242334, US ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-13-3723
  • PATEL S ET AL: "Colony-stimulating factor-1 receptor inhibitors for the treatment of cancer and inflammatory disease", CURRENT TOPICS IN MEDICINAL CHEMISTRY, BENTHAM SCIENCE PUBLISHERS LTD.HILVERSUM, NL, vol. 9, no. 7, 1 January 2009 (2009-01-01) , pages 599-610, XP002570335, ISSN: 1568-0266, DOI: 10.2174/156802609789007327
  • WAGNER CARRIE ET AL: "Evaluation of serum biomarkers associated with radiographic progression in methotrexate-naive rheumatoid arthritis patients treated with methotrexate or golimumab", JOURNAL OF RHEUMATOLOGY, JOURNAL OF RHEUMATOLOGY PUBLISHING COMPANY, CA, vol. 40, no. 5, 30 April 2013 (2013-04-30) , pages 590-598, XP009510622, ISSN: 0315-162X, DOI: 10.3899/JRHEUM.120889
  • RICHARDS D M ET AL: "Immune cell activation by novel hexavalent CD40 agonist APG1233 compared to trimeric formats or agonistic anti-CD40 antibodies", EUROPEAN JOURNAL OF CANCER, vol. 69, 29 November 2016 (2016-11-29), XP029843758, ISSN: 0959-8049, DOI: 10.1016/S0959-8049(16)32893-3
  • KNORR DAVID A ET AL: "Toxicity of an Fc-engineered anti-CD40 antibody is abrogated by intratumoral injection and results in durable antitumor immunity.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 23 10 2018, vol. 115, no. 43, 23 October 2018 (2018-10-23), pages 11048-11053, XP002788203, ISSN: 1091-6490
  
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description

TECHNICAL FIELD



[0001] Means for treating cancer with antibodies that bind colony stimulating factor 1 receptor (CSF1R) in combination with one or more immune stimulating agents.

BACKGROUND



[0002] Colony stimulating factor 1 receptor (referred to herein as CSF1R; also referred to in the art as FMS, FIM2, C-FMS, M-CSF receptor, and CD115) is a single-pass transmembrane receptor with an N-terminal extracellular domain (ECD) and a C-terminal intracellular domain with tyrosine kinase activity. Ligand binding of CSF1 or the interleukin 34 ligand (referred to herein as IL-34; Lin et al., Science 320: 807-11 (2008)) to CSF1R leads to receptor dimerization, upregulation of CSF1R protein tyrosine kinase activity, phosphorylation of CSF1R tyrosine residues, and downstream signaling events. CSF1R activation by CSF1 or IL-34 leads to the trafficking, survival, proliferation, and differentiation of monocytes and macrophages, as well as other monocytic cell lineages such as osteoclasts, dendritic cells, and microglia.

[0003] Many tumor cells or tumor stromal cells have been found to produce CSF1, which activates monocyte/macrophage cells through CSF1R. The level of CSF1 in tumors has been shown to correlate with the level of tumor-associated macrophages (TAMs) in the tumor. Higher levels of TAMs have been found to correlate with poorer patient prognoses in the majority of cancers. In addition, CSF1 has been found to promote tumor growth and progression to metastasis in, for example, human breast cancer xenografts in mice. See, e.g., Paulus et al., Cancer Res. 66: 4349-56 (2006). Further, CSF1R plays a role in osteolytic bone destruction in bone metastasis. See, e.g., Ohno et al., Mol. Cancer Ther. 5: 2634-43 (2006). TAMs promote tumor growth, in part, by suppressing anti-tumor T cell effector function through the release of immunosuppressive cytokines and the expression of T cell inhibitory surface proteins. Thus, antibodies that bind to CSF1R may be useful in methods of treating cancer.

[0004] Some tumor cells may escape detection by the immune system at least in part by suppressing the immune response, for instance by altering the expression of immune modulatory genes. For example, the relative concentrations of both immune stimulatory and immune inhibitory molecules in the body may modulate the adaptive immune response. A relatively high level of expression of inhibitory molecules and/or reduced expression of certain stimulatory molecules may create a checkpoint or switch that down-regulates the adaptive immune response. Agents that counteract this effect by up-regulating the adaptive immune response or by stimulating the innate immune response are potential cancer therapies.

[0005] A combination regimen of agents that modulate the immune response in cancer, such as immune stimulating agents, may increase the depth and durability of the response and may also broaden efficacy to patients who do not respond to a single agent alone.

[0006] WO 2013/132044 A1 discloses combination therapy using antibodies binding to human CSF-1R in combination with a chemotherapeutic agent, radiation and/or cancer immunotherapy.

[0007] WO 2011/140249 discloses anti-CSF-1R antibodies.

SUMMARY



[0008] The invention provides:
  1. (i) an anti-CSFIR antibody for use in a method of treating cancer in a subject, the method comprising administering to the subject the anti-CSFIR antibody and at least one immune stimulating agent;
  2. (ii) at least one immune stimulating agent for use in a method of treating cancer in a subject, the method comprising administering to the subject the at least one immune stimulating agent and an anti-CSFIR antibody; and
  3. (iii) a composition for use in a method of treating cancer, the composition comprising an anti-CSFIR antibody and at least one immune stimulating agent,
wherein in each of (i), (ii) and (iii), the anti-CSFIR antibody binds human CSF1R and is as defined in the claims, and
wherein the at least one immune stimulating agent comprises an agonist anti-CD40 antibody. In the following disclosure, the term "composition of the invention" or "composition for use of the invention" refers to any of (i), (ii) or (iii).

[0009] In some embodiments, the at least one immune stimulating agent further comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, while in some embodiments, the at least one immune stimulating agent further comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule. In some embodiments, the at least one immune stimulating agent further comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, found on immune cells, such as T cells. In some embodiments, the at least one immune stimulating agent further comprises an antagonist of an immune-inhibitory molecule, including a co-inhibitory molecule, found on immune cells, such as T cells. In some embodiments, the at least one immune stimulating agent further comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, found on cells involved in innate immunity, such as NK cells. In some embodiments, the at least one immune stimulating agent further comprises an antagonist of an immune-inhibitory molecule, including a co-inhibitory molecule, found on cells involved in innate immunity, such as NK cells. In some embodiments, the combination enhances the antigen-specific T cell response in the treated subject and/or enhances the innate immunity response in the subject. In some embodiments, the combination results in an improved anti-tumor response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSFIR antibody or immune stimulating agent alone. In some embodiments, the combination results in a synergistic response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSFIR antibody or immune stimulating agent alone.

[0010] In some embodiments, the at least one immune stimulating agent further comprises an antagonist of an inhibitor of the activation of T cells, while in some embodiments, the at least one immune stimulating agent further comprises an agonist of a stimulator of the activation of T cells. In some embodiments, the at least one immune stimulating agent further comprises an antagonist of CTLA4, LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGFβ, VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO. In some embodiments, the at least one immune stimulating agent further comprises an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFNα, STING or a Toll-like receptor agonist such as a TLR2/4 agonist. In some embodiments, the at least one immune stimulating agent further comprises an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. In some embodiments, the at least one immune stimulating agent further comprises an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LTβR, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNFβ, TNFR2, TNFα, 1β2, FAS, FASL, RELT, DR6, TROY, or NGFβ. In some embodiments, the at least one immune stimulating agent further comprises an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGFβ, VEGF. In some embodiments, the at least one immune stimulating agent further comprises an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4. In some embodiments, the at least one immune stimulating agent further comprises an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFNα. In some embodiments, the at least one immune stimulating agent further comprises an antibody. In some embodiments, the at least one immune stimulating agent may further comprise a vaccine, such as a mesothelin-targeting vaccine or attenuated listeria cancer vaccine such as CRS-207. Any one or more of the above antagonists, agonists, and binding agents may be combined with any one or more of the anti-CSFIR antibodies described herein.

[0011] In accordance with the invention, the at least one immune stimulating agent comprises an agonist anti-CD40 antibody, optionally in combination with at least one other immune stimulating agent as listed above. In some embodiments, the anti-CD40 antibody comprises the CDRs of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4. In some embodiments, the anti-CD40 antibody comprises the heavy chain and light chain variable regions of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4. In some embodiments, the anti-CD40 antibody is an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4. In some embodiments, the CD40 agonist is recombinant CD40L. In some embodiments, the at least one immune stimulating agent comprises a CD40 agonist and at least one additional immune stimulating agent from any of those described above. For example, any one or more of the above immune stimulating agents above may be combined with any one or more of the anti-CSFIR antibodies described herein as well as with a CD40 agonist antibody, such as any one of the anti-CD40 antibodies described above.

[0012] In some embodiments, the anti-CSFIR antibody and the at least one immune stimulatory agent are to be administered concurrently or sequentially. In some embodiments, the anti-CSFIR antibody and the at least one immune stimulatory agent are to be administered concurrently. In some embodiments, one or more doses of the at least one immune stimulatory agent are to be administered prior to administering an anti-CSFIR antibody. In some embodiments, the subject received a complete course of therapy with the at least one immune stimulatory agent prior to administration of the anti-CSFIR antibody. In some embodiments, the anti-CSFIR antibody is to be administered during a second course of therapy with the at least one immune stimulatory agent. In some embodiments, the subject received at least one, at least two, at least three, or at least four doses of the at least one immune stimulatory agent prior to administration of the anti-CSFIR antibody. In some embodiments, at least one dose of the at least one immune stimulatory agent is to be administered concurrently with the anti-CSF1R inhibitor. In some embodiments, one or more doses of the anti-CSFIR antibody are to be administered prior to administering at least one immune stimulatory agent. In some embodiments, the subject received at least two, at least three, or at least four doses of the anti-CSF1R antibody prior to administration of at least one immune stimulatory agent. In some embodiments, at least one dose of the anti-CSFIR antibody is to be administered concurrently with the at least one immune stimulatory agent.

[0013] In some embodiments, the cancer is selected from non-small cell lung cancer, melanoma, squamous cell carcinoma of the head and neck, ovarian cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, bladder cancer, endometrial cancer, Hodgkin's lymphoma, lung cancer, glioma, gioblastoma multiforme, colon cancer, breast cancer, bone cancer, skin cancer, uterince cancer, gastric cancer, stomach cancer, lymphoma, lymphocytic leukemia, multiple myeloma, prostate cancer, mesothelioma, and kidney cancer. In some embodiments, the cancer is recurrent or progressive after a therapy selected from surgery, chemotherapy, radiation therapy, or a combination thereof.

[0014] In some embodiments of the compositions for use of the invention, the at least one immune stimulating agent further comprises an antagonist of an inhibitor of the activation of T cells, while in some embodiments, the at least one immune stimulating agent further comprises an agonist of a stimulator of the activation of T cells. In some embodiments, the at least one immune stimulating agent further comprises an antagonist of CTLA4, LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGFβ, VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO. In some embodiments, the at least one immune stimulating agent further comprises an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFNα, STING, or a Toll-like receptor agonist such as a TLR2/4 agonist. In some embodiments, the at least one immune stimulating agent further comprises an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. In some embodiments, the at least one immune stimulating agent further comprises an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LTβR, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNFβ, TNFR2, TNFα, 1β2, FAS, FASL, RELT, DR6, TROY, or NGFβ. In some embodiments, the at least one immune stimulating agent further comprises an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGFβ, VEGF. In some embodiments, the at least one immune stimulating agent further comprises an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFNα. In some embodiments, the at least one immune stimulating agent further comprises an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4. In some embodiments, the at least one immune stimulating agent further comprises an antibody. In some embodiments, the at least one immune stimulating agent may further comprise a vaccine, such as a mesothelin-targeting vaccine or attenuated listeria cancer vaccine such as CRS-207.

[0015] Subject to the requirements of the claims, the compositions comprise any one or more of the above antagonists, agonists, and binding agents combined with any one or more of the anti-CSFIR antibodies described herein. The compositions may include each therapeutic agent in a separate container or compartment or alternatively, may include two or more of the therapeutic agents mixed together.

[0016] In accordance with the invention, the compositions comprise an anti-CSFIR antibody and an agonist anti-CD40 antibody, optionally along with at least one other immune stimulating agent as listed above. In some embodiments, the anti-CD40 antibody comprises the CDRs of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4. In some embodiments, the anti-CD40 antibody comprises the heavy chain and light chain variable regions of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4. In some embodiments, the anti-CD40 antibody is an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4. In some embodiments, the CD40 agonist is recombinant CD40L. In some embodiments, the compositions comprise any one or more of the above immune stimulating agents above combined with both any one or more of the anti-CSF1R antibodies described herein as well as with a CD40 agonist antibody, such as any one of the anti-CD40 antibodies described above. The compositions may include each therapeutic agent in a separate container or compartment or alternatively, may include two or more of the therapeutic agents mixed together.

[0017] In the compositions of the invention, the antibody heavy chain and the antibody light chain of the anti-CSFIR antibody have the structure as defined in the claims.

[0018] The remainder of this paragraph is subject to the requirements of the claims concerning the structure of the anti-CSFIR antibody. In any of the compositions of the invention, the anti-CSFIR antibody heavy chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 39. In any of the compositions of the invention, the anti-CSFIR antibody light chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 46. In any of the compositions of the invention, the anti-CSFIR antibody heavy chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 39, and the anti-CSFIR antibody light chain may comprise a sequence that is at least 90%, at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 46.

[0019] The remainder of this paragraph is subject to the requirements of the claims concerning the structure of the anti-CSFIR antibody. In any of the compositions of the invention, the anti-CSFIR antibody may comprise a heavy chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 39 and a light chain comprising a sequence that is at least 95%, at least 97%, at least 99%, or 100% identical to SEQ ID NO: 46.

[0020] In any of the compositions of the invention, the anti-CSFIR antibody may comprise a heavy chain comprising a heavy chain (HC) CDR1 having the sequence of SEQ ID NO: 15, an HC CDR2 having the sequence of SEQ ID NO: 16, and an HC CDR3 having the sequence of SEQ ID NO: 17, and a light chain comprising a light chain (LC) CDR1 having the sequence of SEQ ID NO: 18, a LC CDR2 having the sequence of SEQ ID NO: 19, and a LC CDR3 having the sequence of SEQ ID NO: 20.

[0021] In any of the compositions of the invention, the anti-CSFIR antibody may comprise a heavy chain comprising a sequence of SEQ ID NO: 53 and a light chain comprising a sequence of SEQ ID NO: 60.

[0022] In any of the compositions of the invention, the anti-CSFIR antibody may be a humanized antibody. In any of the compositions or methods described herein, the anti-CSFIR antibody may be selected from a Fab, an Fv, an scFv, a Fab', and a (Fab')2. In any of the compositions or methods described herein, the anti-CSFIR antibody may be a chimeric antibody. In any of the compositions or methods described herein, the anti-CSFIR antibody may be selected from an IgA, an IgG, and an IgD. In any of the compositions or methods described herein, the anti-CSFIR antibody may be an IgG. In any of the methods described herein, the antibody may be an IgG1 or IgG2.

[0023] In any of the compositions of the invention, the anti-CSFIR antibody binds to human CSF1R and may bind to cynomolgus CSF1R. In any of the compositions of the invention, the anti-CSFIR antibody may block ligand binding to CSF1R. In any of the compositions of the invention, the anti-CSFIR antibody may block binding of CSF1 and/or IL-34 to CSF1R. In any of the compositions of the invention, the anti-CSFIR antibody may block binding of both CSF1 and IL-34 to CSF1R. In any of the compositions of the invention, the anti-CSFIR antibody may inhibit ligand-induced CSF1R phosphorylation. In any of the compositions of the invention, the anti-CSFIR antibody may inhibit CSF1- and/or IL-34-induced CSF1R phosphorylation. In any of the compositions of the invention, the anti-CSFIR antibody may bind to human CSF1R with an affinity (KD) of less than 1 nM. In any of the compositions of the invention, the anti-CSFIR antibody may inhibit monocyte proliferation and/or survival responses in the presence of CSF1 or IL-34.

BRIEF DESCRIPTION OF THE FIGURES



[0024] 

FIG. 1A-C show an alignment of the humanized heavy chain variable regions for each of humanized antibodies huAb1 to huAb16, as discussed in Example 1. Boxed residues are amino acids in the human acceptor sequence that were changed back to the corresponding mouse residue.

FIG. 2A-C show an alignment of the humanized light chain variable regions for each of humanized antibodies huAb1 to huAb16, as discussed in Example 1. Boxed amino acids are residues in the human acceptor sequence that were changed back to the corresponding mouse residue.

FIG. 3 shows that the combination of anti-CSF1R antibody and anti-CD40 antibody demonstrate greater tumor growth suppression in an MC38 tumor mouse model than either therapy alone.

FIG. 4A-B shows the tumor volume of individual mice at (Fig. 4A) day 11 and (Fig. 4B) day 13. The combination of anti-CSFIR antibody and anti-CD40 antibody performed significantly better than either therapy alone at both time points.

FIG. 5 shows body weight in mice used in the study.


DETAILED DESCRIPTION



[0025] Tumor-associated macrophages (TAMs) are implicated in the pathogenesis of many cancers, and correlate with poor prognosis. TAMs can suppress an anti-tumor responses through multiple mechanisms. TAMs express anti-inflammatory cytokines such as TGFβ and IL-10 which acts to suppress the ability of intratumoral dendritic cells to stimulate cytotoxic T cell responses (Ruffell et al., 2014, Cancer Cell). TAMs also express chemokines which recruit immunosuppressive regulatory T cells into tumors (Curiel et al., 2004, Nature Med.; Mizukami et al., 2008, Int. J. Cancer), Moreover, TAMs express the ligands for the T cell inhibitor receptors PD-1 and CTLA-4 which act to directly inhibit T cell activation and funnction. Inhibition of CSF1R can reduce immunosuppressive TAMs in mouse models and human tumors. See, e.g., Ries et al., 2014, Cancer Cell, 25: 846-859; Pyontech et al., 2013, Nature Med., 19: 1264-1272; and Zhu et al., 2014, Cancer Res., 74: 5057-5069. In contrast to CSF1R blockade which acts by reducing immunosuppression, immune stimulating agents work by stimulating an immune response.

[0026] The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.

Definitions



[0027] Unless otherwise defined, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.

[0028] Exemplary techniques used in connection with recombinant DNA, oligonucleotide synthesis, tissue culture and transformation (e.g., electroporation, lipofection), enzymatic reactions, and purification techniques are known in the art. Many such techniques and procedures are described, e.g., in Sambrook et al. Molecular Cloning: A Laboratory Manual (2nd ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), among other places. In addition, exemplary techniques for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, and delivery, and treatment of patients are also known in the art.

[0029] In this application, the use of "or" means "and/or" unless stated otherwise. In the context of a multiple dependent claim, the use of "or" refers back to more than one preceding independent or dependent claim in the alternative only. Also, terms such as "element" or "component" encompass both elements and components comprising one unit and elements and components that comprise more than one subunit unless specifically stated otherwise.

[0030] As utilized in accordance with the present disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings:

[0031] The terms "nucleic acid molecule" and "polynucleotide" may be used interchangeably, and refer to a polymer of nucleotides. Such polymers of nucleotides may contain natural and/or non-natural nucleotides, and include, but are not limited to, DNA, RNA, and PNA. "Nucleic acid sequence" refers to the linear sequence of nucleotides that comprise the nucleic acid molecule or polynucleotide.

[0032] The terms "polypeptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues, and are not limited to a minimum length. Such polymers of amino acid residues may contain natural or non-natural amino acid residues, and include, but are not limited to, peptides, oligopeptides, dimers, trimers, and multimers of amino acid residues. Both full-length proteins and fragments thereof are encompassed by the definition. The terms also include post-expression modifications of the polypeptide, for example, glycosylation, sialylation, acetylation, phosphorylation, and the like. Furthermore, for purposes of the present invention, a "polypeptide" refers to a protein which includes modifications, such as deletions, additions, and substitutions (generally conservative in nature), to the native sequence, as long as the protein maintains the desired activity. These modifications may be deliberate, as through site-directed mutagenesis, or may be accidental, such as through mutations of hosts which produce the proteins or errors due to PCR amplification.

[0033] The term "CSF1R" refers herein to the full-length CSF1R, which includes the N-terminal ECD, the transmembrane domain, and the intracellular tyrosine kinase domain, with or without an N-terminal leader sequence. In some embodiments, the CSF1R is a human CSF1R having the amino acid sequence of SEQ ID NO: 1 or SEQ ID NO: 2.

[0034] The term "immune stimulating agent" as used herein refers to a molecule that stimulates the immune system by either acting as an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, or acting as an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule. An immune stimulating agent may be a biologic, such as an antibody or antibody fragment, other protein, or vaccine, or may be a small molecule drug. An "immune stimulatory molecule" includes a receptor or ligand that acts to enhance, stimulate, induce, or otherwise "turn-on" an immune response. Immune stimulatory molecules as defined herein include co-stimulatory molecules. An "immune inhibitory molecule" includes a receptor or ligand that acts to reduce, inhibit, suppress, or otherwise "turn-off" an immune response. Immune inhibitory molecules as defined herein include co-inhibitory molecules. Such immune stimulatory and immune inhibitory molecules may be, for example, receptors or ligands found on immune cells such as a T cells, or found on cells involved in innate immunity such as NK cells.

[0035] The terms "B-cell surface antigen CD40" and "CD40" refer herein to the full-length CD40, which includes the N-terminal ECD, the transmembrane domain, and the intracellular domain, with or without an N-terminal leader sequence. In some embodiments, the CD40 is a human CD40 having the amino acid sequence of SEQ ID NO: 96 (precursor, with signal sequence) or SEQ ID NO: 97 (mature, without signal sequence).

[0036] The term "CD40 agonist" refers to a moiety that interacts with CD40 and enhances CD40 activity. Nonlimiting exemplary CD40 activities include signaling through CD40, enhancement of antigen presenting activity, induction of proinflammatory cytokines, and induction of tumorcidal activity. In some embodiments, a CD40 agonist is an anti-CD40 antibody.

[0037] The term "anti-CD40 antibody" refers to an antibody that specifically binds to CD40. Unless specifically indicated otherwise, the term "anti-CD40 antibody" as used herein refers to an anti-CD40 agonist antibody.

[0038] With reference to anti-CSFIR antibodies the term "blocks binding of" a ligand, such as CSF1 and/or IL-34, and grammatical variants thereof, are used to refer to the ability to inhibit the interaction between CSF1R and a CSF1R ligand, such as CSF1 and/or IL-34. Such inhibition may occur through any mechanism, including direct interference with ligand binding, e.g., because of overlapping binding sites on CSF1R, and/or conformational changes in CSF1R induced by the antibody that alter ligand affinity, etc. Antibodies and antibody fragments referred to as "functional" are characterized by having such properties.

[0039] The term "antibody" as used herein refers to a molecule comprising at least complementarity-determining region (CDR) 1, CDR2, and CDR3 of a heavy chain and at least CDR1, CDR2, and CDR3 of a light chain, wherein the molecule is capable of binding to antigen. The term antibody includes, but is not limited to, fragments that are capable of binding antigen, such as Fv, single-chain Fv (scFv), Fab, Fab', and (Fab')2. The term antibody also includes, but is not limited to, chimeric antibodies, humanized antibodies, and antibodies of various species such as mouse, human, cynomolgus monkey, etc.

[0040] In some embodiments, an antibody comprises a heavy chain variable region and a light chain variable region. In some embodiments, an antibody comprises at least one heavy chain comprising a heavy chain variable region and at least a portion of a heavy chain constant region, and at least one light chain comprising a light chain variable region and at least a portion of a light chain constant region. In some embodiments, an antibody comprises two heavy chains, wherein each heavy chain comprises a heavy chain variable region and at least a portion of a heavy chain constant region, and two light chains, wherein each light chain comprises a light chain variable region and at least a portion of a light chain constant region. As used herein, a single-chain Fv (scFv), or any other antibody that comprises, for example, a single polypeptide chain comprising all six CDRs (three heavy chain CDRs and three light chain CDRs) is considered to have a heavy chain and a light chain. In some such embodiments, the heavy chain is the region of the antibody that comprises the three heavy chain CDRs and the light chain in the region of the antibody that comprises the three light chain CDRs.

[0041] The term "heavy chain variable region" as used herein refers to a region comprising heavy chain CDR1, framework (FR) 2, CDR2, FR3, and CDR3. In some embodiments, a heavy chain variable region also comprises at least a portion of an FR1 and/or at least a portion of an FR4. In some embodiments, a heavy chain CDR1 corresponds to Kabat residues 26 to 35; a heavy chain CDR2 corresponds to Kabat residues 50 to 65; and a heavy chain CDR3 corresponds to Kabat residues 95 to 102. See, e.g., Kabat Sequences of Proteins of Immunological Interest (1987 and 1991, NIH, Bethesda, Md.); and Figure 1. In some embodiments, a heavy chain CDR1 corresponds to Kabat residues 31 to 35; a heavy chain CDR2 corresponds to Kabat residues 50 to 65; and a heavy chain CDR3 corresponds to Kabat residues 95 to 102. See id.

[0042] The term "heavy chain constant region" as used herein refers to a region comprising at least three heavy chain constant domains, CH1, CH2, and CH3. Nonlimiting exemplary heavy chain constant regions include γ, δ, and α. Nonlimiting exemplary heavy chain constant regions also include ε and µ. Each heavy constant region corresponds to an antibody isotype. For example, an antibody comprising a γ constant region is an IgG antibody, an antibody comprising a δ constant region is an IgD antibody, and an antibody comprising an α constant region is an IgA antibody. Further, an antibody comprising a µ constant region is an IgM antibody, and an antibody comprising an ε constant region is an IgE antibody. Certain isotypes can be further subdivided into subclasses. For example, IgG antibodies include, but are not limited to, IgG1 (comprising a γ1 constant region), IgG2 (comprising a γ2 constant region), IgG3 (comprising a γ3 constant region), and IgG4 (comprising a γ4 constant region) antibodies; IgA antibodies include, but are not limited to, IgA1 (comprising an α1 constant region) and IgA2 (comprising an α2 constant region) antibodies; and IgM antibodies include, but are not limited to, IgM1 and IgM2.

[0043] In some embodiments, a heavy chain constant region comprises one or more mutations (or substitutions), additions, or deletions that confer a desired characteristic on the antibody. A nonlimiting exemplary mutation is the S241P mutation in the IgG4 hinge region (between constant domains CH1 and CH2), which alters the IgG4 motif CPSCP to CPPCP, which is similar to the corresponding motif in IgG1. That mutation, in some embodiments, results in a more stable IgG4 antibody. See, e.g., Angal et al., Mol. Immunol. 30: 105-108 (1993); Bloom et al., Prot. Sci. 6: 407-415 (1997); Schuurman et al., Mol. Immunol. 38: 1-8 (2001).

[0044] The term "heavy chain" as used herein refers to a polypeptide comprising at least a heavy chain variable region, with or without a leader sequence. In some embodiments, a heavy chain comprises at least a portion of a heavy chain constant region. The term "full-length heavy chain" as used herein refers to a polypeptide comprising a heavy chain variable region and a heavy chain constant region, with or without a leader sequence.

[0045] The term "light chain variable region" as used herein refers to a region comprising light chain CDR1, framework (FR) 2, CDR2, FR3, and CDR3. In some embodiments, a light chain variable region also comprises an FR1 and/or an FR4. In some embodiments, a light chain CDR1 corresponds to Kabat residues 24 to 34; a light chain CDR2 corresponds to Kabat residues 50 to 56; and a light chain CDR3 corresponds to Kabat residues 89 to 97. See, e.g., Kabat Sequences of Proteins of Immunological Interest (1987 and 1991, NIH, Bethesda, Md.); and Figure 1.

[0046] The term "light chain constant region" as used herein refers to a region comprising a light chain constant domain, CL. Nonlimiting exemplary light chain constant regions include λ and κ.

[0047] The term "light chain" as used herein refers to a polypeptide comprising at least a light chain variable region, with or without a leader sequence. In some embodiments, a light chain comprises at least a portion of a light chain constant region. The term "full-length light chain" as used herein refers to a polypeptide comprising a light chain variable region and a light chain constant region, with or without a leader sequence.

[0048] A "chimeric antibody" as used herein refers to an antibody comprising at least one variable region from a first species (such as mouse, rat, cynomolgus monkey, etc.) and at least one constant region from a second species (such as human, cynomolgus monkey, etc.). In some embodiments, a chimeric antibody comprises at least one mouse variable region and at least one human constant region. In some embodiments, a chimeric antibody comprises at least one cynomolgus variable region and at least one human constant region. In some embodiments, a chimeric antibody comprises at least one rat variable region and at least one mouse constant region. In some embodiments, all of the variable regions of a chimeric antibody are from a first species and all of the constant regions of the chimeric antibody are from a second species.

[0049] A "humanized antibody" as used herein refers to an antibody in which at least one amino acid in a framework region of a non-human variable region has been replaced with the corresponding amino acid from a human variable region. In some embodiments, a humanized antibody comprises at least one human constant region or fragment thereof. In some embodiments, a humanized antibody is a Fab, an scFv, a (Fab')2, etc.

[0050] A "CDR-grafted antibody" as used herein refers to a humanized antibody in which the complementarity determining regions (CDRs) of a first (non-human) species have been grafted onto the framework regions (FRs) of a second (human) species.

[0051] A "human antibody" as used herein refers to antibodies produced in humans, antibodies produced in non-human animals that comprise human immunoglobulin genes, such as XenoMouse®, and antibodies selected using in vitro methods, such as phage display, wherein the antibody repertoire is based on a human immunoglobulin sequences.

[0052] The term "leader sequence" refers to a sequence of amino acid residues located at the N terminus of a polypeptide that facilitates secretion of a polypeptide from a mammalian cell. A leader sequence may be cleaved upon export of the polypeptide from the mammalian cell, forming a mature protein. Leader sequences may be natural or synthetic, and they may be heterologous or homologous to the protein to which they are attached. Exemplary leader sequences include, but are not limited to, antibody leader sequences, such as, for example, the amino acid sequences of SEQ ID NOs: 3 and 4, which correspond to human light and heavy chain leader sequences, respectively. Nonlimiting exemplary leader sequences also include leader sequences from heterologous proteins. In some embodiments, an antibody lacks a leader sequence. In some embodiments, an antibody comprises at least one leader sequence, which may be selected from native antibody leader sequences and heterologous leader sequences.

[0053] The term "vector" is used to describe a polynucleotide that may be engineered to contain a cloned polynucleotide or polynucleotides that may be propagated in a host cell. A vector may include one or more of the following elements: an origin of replication, one or more regulatory sequences (such as, for example, promoters and/or enhancers) that regulate the expression of the polypeptide of interest, and/or one or more selectable marker genes (such as, for example, antibiotic resistance genes and genes that may be used in colorimetric assays, e.g., β-galactosidase). The term "expression vector" refers to a vector that is used to express a polypeptide of interest in a host cell.

[0054] A "host cell" refers to a cell that may be or has been a recipient of a vector or isolated polynucleotide. Host cells may be prokaryotic cells or eukaryotic cells. Exemplary eukaryotic cells include mammalian cells, such as primate or non-primate animal cells; fungal cells, such as yeast; plant cells; and insect cells. Nonlimiting exemplary mammalian cells include, but are not limited to, NSO cells, PER.C6® cells (Crucell), and 293 and CHO cells, and their derivatives, such as 293-6E and DG44 cells, respectively.

[0055] The term "isolated" as used herein refers to a molecule that has been separated from at least some of the components with which it is typically found in nature. For example, a polypeptide is referred to as "isolated" when it is separated from at least some of the components of the cell in which it was produced. Where a polypeptide is secreted by a cell after expression, physically separating the supernatant containing the polypeptide from the cell that produced it is considered to be "isolating" the polypeptide. Similarly, a polynucleotide is referred to as "isolated" when it is not part of the larger polynucleotide (such as, for example, genomic DNA or mitochondrial DNA, in the case of a DNA polynucleotide) in which it is typically found in nature, or is separated from at least some of the components of the cell in which it was produced, e.g., in the case of an RNA polynucleotide. Thus, a DNA polynucleotide that is contained in a vector inside a host cell may be referred to as "isolated" so long as that polynucleotide is not found in that vector in nature.

[0056] The term "elevated level" means a higher level of a protein in a particular tissue of a subject relative to the same tissue in a control, such as an individual or individuals who are not suffering from cancer or other condition described herein. The elevated level may be the result of any mechanism, such as increased expression, increased stability, decreased degradation, increased secretion, decreased clearance, etc., of the protein.

[0057] The term "reduce" or "reduces" means to lower the level of a protein in a particular tissue of a subject by at least 10%. In some embodiments, an agent, such as an antibody that binds CSF1R, reduces the level of a protein in a particular tissue of a subject by at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90%. In some embodiments, the level of a protein is reduced relative to the level of the protein prior to contacting with an agent, such as an antibody that binds CSF1R.

[0058] The term "resistant," when used in the context of resistance to a therapeutic agent, means a decreased response or lack of response to a standard dose of the therapeutic agent, relative to the subject's response to the standard dose of the therapeutic agent in the past, or relative to the expected response of a similar subject with a similar disorder to the standard dose of the therapeutic agent. Thus, in some embodiments, a subject may be resistant to therapeutic agent although the subject has not previously been given the therapeutic agent, or the subject may develop resistance to the therapeutic agent after having responded to the agent on one or more previous occasions.

[0059] The terms "subject" and "patient" are used interchangeably herein to refer to a human. In some embodiments, methods of treating other mammals, including, but not limited to, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets, are also provided.

[0060] The term "sample," as used herein, refers to a composition that is obtained or derived from a subject that contains a cellular and/or other molecular entity that is to be characterized, quantitated, and/or identified, for example based on physical, biochemical, chemical and/or physiological characteristics. An exemplary sample is a tissue sample.

[0061] The term "tissue sample" refers to a collection of similar cells obtained from a tissue of a subject. The source of the tissue sample may be solid tissue as from a fresh, frozen and/or preserved organ or tissue sample or biopsy or aspirate; blood or any blood constituents; bodily fluids such as cerebral spinal fluid, amniotic fluid, peritoneal fluid, synovial fluid, or interstitial fluid; cells from any time in gestation or development of the subject. In some embodiments, a tissue sample is a synovial biopsy tissue sample and/or a synovial fluid sample. In some embodiments, a tissue sample is a synovial fluid sample. The tissue sample may also be primary or cultured cells or cell lines. Optionally, the tissue sample is obtained from a disease tissue/organ. The tissue sample may contain compounds that are not naturally intermixed with the tissue in nature such as preservatives, anticoagulants, buffers, fixatives, nutrients, antibiotics, or the like. A "control sample" or "control tissue", as used herein, refers to a sample, cell, or tissue obtained from a source known, or believed, not to be afflicted with the disease for which the subject is being treated.

[0062] For the purposes herein a "section" of a tissue sample means a part or piece of a tissue sample, such as a thin slice of tissue or cells cut from a solid tissue sample.

[0063] The term "cancer" is used herein to refer to a group of cells that exhibit abnormally high levels of proliferation and growth. A cancer may be benign (also referred to as a benign tumor), pre-malignant, or malignant. Cancer cells may be solid cancer cells or leukemic cancer cells. The term "cancer growth" is used herein to refer to proliferation or growth by a cell or cells that comprise a cancer that leads to a corresponding increase in the size or extent of the cancer.

[0064] Examples of cancer include but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular nonlimiting examples of such cancers include squamous cell cancer, small-cell lung cancer, pituitary cancer, esophageal cancer, astrocytoma, soft tissue sarcoma, non-small cell lung cancer (including squamous cell non-small cell lung cancer), adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, renal cell carcinoma, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, brain cancer, endometrial cancer, testis cancer, cholangiocarcinoma, gallbladder carcinoma, gastric cancer, melanoma, and various types of head and neck cancer (including squamous cell carcinoma of the head and neck).

[0065] The term "recurrent cancer" refers to a cancer that has returned after a previous treatment regimen, following which there was a period of time during which the cancer could not be detected.

[0066] The term "progressive cancer" is a cancer that has increased in size or tumor spread since the beginning of a treatment regimen. In certain embodiments, a progressive cancer is a cancer that has incrased in size or tumor spread by at least 10%, at least 20%, at least 30%, at least 40%, or at least 50% since the beginning of a treatment regimen.

[0067] A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer. Examples of chemotherapeutic agents include, but are not limited to, alkylating agents such as thiotepa and Cytoxan® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gamma1I and calicheamicin omegaI1 (see, e.g., Agnew, Chem Intl. Ed. Engl., 33: 183-186 (1994)); dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, Adriamycin® doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2- ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., Taxol® paclitaxel (Bristol- Myers Squibb Oncology, Princeton, N.J.), Abraxane® Cremophor-free, albumin-engineered nanoparticle formulation of paclitaxel (American Pharmaceutical Partners, Schaumberg, Illinois), and Taxotere® doxetaxel (Rhône- Poulenc Rorer, Antony, France); chloranbucil; Gemzar® gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; Navelbine® vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (Camptosar, CPT-11) (including the treatment regimen of irinotecan with 5-FU and leucovorin); topoisomerase inhibitor RFS 2000; difluorometlhylornithine (DMFO); retinoids such as retinoic acid; capecitabine; combretastatin; leucovorin (LV); oxaliplatin, including the oxaliplatin treatment regimen (FOLFOX); inhibitors of PKC-alpha, Raf, H-Ras, EGFR (e.g., erlotinib (Tarceva®)) and VEGF-A that reduce cell proliferation and pharmaceutically acceptable salts, acids or derivatives of any of the above.

[0068] Further nonlimiting exemplary chemotherapeutic agents include anti-hormonal agents that act to regulate or inhibit hormone action on cancers such as anti-estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including Nolvadex® tamoxifen), raloxifene, droloxifene, 4-hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and Fareston® toremifene; aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, Megase® megestrol acetate, Aromasin® exemestane, formestanie, fadrozole, Rivisor® vorozole, Femara® letrozole, and Arimidex® anastrozole; and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in abherant cell proliferation, such as, for example, PKC-alpha, Ralf and H-Ras; ribozymes such as a VEGF expression inhibitor (e.g., Angiozyme® ribozyme) and a HER2 expression inhibitor; vaccines such as gene therapy vaccines, for example, Allovectin® vaccine, Leuvectin® vaccine, and Vaxid® vaccine; Proleukin® rIL-2; Lurtotecan® topoisomerase 1 inhibitor; Abarelix® rmRH; and pharmaceutically acceptable salts, acids or derivatives of any of the above.

[0069] An "anti-angiogenesis agent" or "angiogenesis inhibitor" refers to a small molecular weight substance, a polynucleotide (including, e.g., an inhibitory RNA (RNAi or siRNA)), a polypeptide, an isolated protein, a recombinant protein, an antibody, or conjugates or fusion proteins thereof, that inhibits angiogenesis, vasculogenesis, or undesirable vascular permeability, either directly or indirectly. It should be understood that the anti-angiogenesis agent includes those agents that bind and block the angiogenic activity of the angiogenic factor or its receptor. For example, an anti-angiogenesis agent is an antibody or other antagonist to an angiogenic agent, e.g., antibodies to VEGF-A (e.g., bevacizumab (Avastin®)) or to the VEGF-A receptor (e.g., KDR receptor or Flt-1 receptor), anti-PDGFR inhibitors such as Gleevec® (Imatinib Mesylate), small molecules that block VEGF receptor signaling (e.g., PTK787/ZK2284, SU6668, Sutent®/SU11248 (sunitinib malate), AMG706, or those described in, e.g., international patent application WO 2004/113304). Anti-angiogensis agents also include native angiogenesis inhibitors , e.g., angiostatin, endostatin, etc. See, e.g., Klagsbrun and D'Amore (1991) Annu. Rev. Physiol. 53:217-39; Streit and Detmar (2003) Oncogene 22:3172-3179 (e.g., Table 3 listing anti-angiogenic therapy in malignant melanoma); Ferrara & Alitalo (1999) Nature Medicine 5(12): 1359-1364; Tonini et al. (2003) Oncogene 22:6549-6556 (e.g., Table 2 listing known anti-angiogenic factors); and, Sato (2003) Int. J. Clin. Oncol. 8:200-206 (e.g., Table 1 listing anti-angiogenic agents used in clinical trials).

[0070] A "growth inhibitory agent" as used herein refers to a compound or composition that inhibits growth of a cell (such as a cell expressing VEGF) either in vitro or in vivo. Thus, the growth inhibitory agent may be one that significantly reduces the percentage of cells (such as a cell expressing VEGF) in S phase. Examples of growth inhibitory agents include, but are not limited to, agents that block cell cycle progression (at a place other than S phase), such as agents that induce G1 arrest and M-phase arrest. Classical M-phase blockers include the vincas (vincristine and vinblastine), taxanes, and topoisomerase II inhibitors such as doxorubicin, epirubicin, daunorubicin, etoposide, and bleomycin. Those agents that arrest G1 also spill over into S-phase arrest, for example, DNA alkylating agents such as tamoxifen, prednisone, dacarbazine, mechlorethamine, cisplatin, methotrexate, 5-fluorouracil, and ara-C. Further information can be found in Mendelsohn and Israel, eds., The Molecular Basis of Cancer, Chapter 1, entitled "Cell cycle regulation, oncogenes, and antineoplastic drugs" by Murakami et al. (W.B. Saunders, Philadelphia, 1995), e.g., p. 13. The taxanes (paclitaxel and docetaxel) are anticancer drugs both derived from the yew tree. Docetaxel (Taxotere®, Rhone-Poulenc Rorer), derived from the European yew, is a semisynthetic analogue of paclitaxel (Taxol®, Bristol-Myers Squibb). Paclitaxel and docetaxel promote the assembly of microtubules from tubulin dimers and stabilize microtubules by preventing depolymerization, which results in the inhibition of mitosis in cells.

[0071] The term "anti-neoplastic composition" refers to a composition useful in treating cancer comprising at least one active therapeutic agent. Examples of therapeutic agents include, but are not limited to, e.g., chemotherapeutic agents, growth inhibitory agents, cytotoxic agents, agents used in radiation therapy, anti-angiogenesis agents, cancer immunotherapeutic agents, apoptotic agents, anti-tubulin agents, and other-agents to treat cancer, such as anti-HER-2 antibodies, anti-CD20 antibodies, an epidermal growth factor receptor (EGFR) antagonist (e.g., a tyrosine kinase inhibitor), HER1/EGFR inhibitor (e.g., erlotinib (Tarceva®), platelet derived growth factor inhibitors (e.g., Gleevec® (Imatinib Mesylate)), a COX-2 inhibitor (e.g., celecoxib), interferons, CTLA4 inhibitors (e.g., anti-CTLA antibody ipilimumab (YERVOY®)), TIM3 inhibitors (e.g., anti-TIM3 antibodies), cytokines, antagonists (e.g., neutralizing antibodies) that bind to one or more of the following targets ErbB2, ErbB3, ErbB4, PDGFR-beta, BlyS, APRIL, BCMA, CTLA4, TIM3, or VEGF receptor(s), TRAIL/Apo2, and other bioactive and organic chemical agents, etc. Combinations thereof are also included in the invention.

[0072] An agent "antagonizes" factor activity when the agent neutralizes, blocks, inhibits, abrogates, reduces, and/or interferes with the activity of the factor, including its binding to one or more receptors when the factor is a ligand.

[0073] "Treatment," as used herein, refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) the targeted pathologic condition or disorder. In certain embodiments, the term "treatment" covers any administration or application of a therapeutic for disease in a mammal, including a human, and includes inhibiting or slowing the disease or progression of the disease; partially or fully relieving the disease, for example, by causing regression, or restoring or repairing a lost, missing, or defective function; stimulating an inefficient process; or causing the disease plateau to have reduced severity. The term "treatment" also includes reducing the severity of any phenotypic characteristic and/or reducing the incidence, degree, or likelihood of that characteristic. Those in need of treatment include those already with the disorder as well as those prone to have the disorder or those in whom the disorder is to be prevented.

[0074] The term "effective amount" or "therapeutically effective amount" refers to an amount of a drug effective to treat a disease or disorder in a subject. In certain embodiments, an effective amount refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result. A therapeutically effective amount of an anti-CSFIR antibody and/or an immune stimulating agent of the invention may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibodies to elicit a desired response in the individual. A therapeutically effective amount encompasses an amount in which any toxic or detrimental effects of the antibody or antibodies are outweighed by the therapeutically beneficial effects. In some embodiments, the expression "effective amount" refers to an amount of the antibody that is effective for treating the cancer.

[0075] A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount would be less than the therapeutically effective amount.

[0076] Administration "in combination with" one or more further therapeutic agents includes simultaneous (concurrent) and consecutive (sequential) administration in any order.

[0077] A "pharmaceutically acceptable carrier" refers to a non-toxic solid, semisolid, or liquid filler, diluent, encapsulating material, formulation auxiliary, or carrier conventional in the art for use with a therapeutic agent that together comprise a "pharmaceutical composition" for administration to a subject. A pharmaceutically acceptable carrier is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. The pharmaceutically acceptable carrier is appropriate for the formulation employed. For example, if the therapeutic agent is to be administered orally, the carrier may be a gel capsule. If the therapeutic agent is to be administered subcutaneously, the carrier ideally is not irritable to the skin and does not cause injection site reaction.

Anti-CSFIR Antibodies



[0078] Anti-CSF1R antibodies include, but are not limited to, humanized antibodies, chimeric antibodies, mouse antibodies, human antibodies, and antibodies comprising the heavy chain and/or light chain CDRs discussed herein. The anti-CSFIR antibodies that are included in the compositions for use according to the invention (or with which the compositions for use according to the invention are to be used) are as defined in the claims. The remaining disclosure of this section is subject to the requirements of the claims.

Exemplary Humanized Antibodies



[0079] In some embodiments, humanized antibodies that bind CSF1R are provided. Humanized antibodies are useful as therapeutic molecules because humanized antibodies reduce or eliminate the human immune response to non-human antibodies (such as the human anti-mouse antibody (HAMA) response), which can result in an immune response to an antibody therapeutic, and decreased effectiveness of the therapeutic.

[0080] Nonlimiting exemplary humanized antibodies include huAb1 through huAb16, described herein. Nonlimiting exemplary humanized antibodies also include antibodies comprising a heavy chain variable region of an antibody selected from huAb1 to huAb16 and/or a light chain variable region of an antibody selected from huAb1 to huAb16. Nonlimiting exemplary humanized antibodies include antibodies comprising a heavy chain variable region selected from SEQ ID NOs: 39 to 45 and/or a light chain variable region selected from SEQ ID NOs: 46 to 52. Exemplary humanized antibodies also include, but are not limited to, humanized antibodies comprising heavy chain CDR1, CDR2, and CDR3, and/or light chain CDR1, CDR2, and CDR3 of an antibody selected from 0301, 0302, and 0311.

[0081] In some embodiments, a humanized anti-CSFIR antibody comprises heavy chain CDR1, CDR2, and CDR3 and/or a light chain CDR1, CDR2, and CDR3 of an antibody selected from 0301, 0302, and 0311. Nonlimiting exemplary humanized anti-CSFIR antibodies include antibodies comprising sets of heavy chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 15, 16, and 17; SEQ ID NOs: 21, 22, and 23; and SEQ ID NOs: 27, 28, and 29. Nonlimiting exemplary humanized anti-CSFIR antibodies also include antibodies comprising sets of light chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 18, 19, and 20; SEQ ID NOs: 24, 25, and 26; and SEQ ID NOs: 30, 31, and 32.

[0082] Nonlimiting exemplary humanized anti-CSFIR antibodies include antibodies comprising the sets of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 in Table 1 (SEQ ID NOs shown; see Table 8 for sequences). Each row of Table 1 shows the heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 of an exemplary antibody.
Table 1: Heavy chain and light chain CDRs
 Heavy chainLight chain
AbCDR1 SEQ IDCDR2 SEQ IDCDR3 SEQ IDCDR1 SEQ IDCDR2 SEQ IDCDR3 SEQ ID
0301 15 16 17 18 19 20
0302 21 22 23 24 25 26
0311 27 28 29 30 31 32

Further exemplary humanized antibodies



[0083] In some embodiments, a humanized anti-CSFIR antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, and wherein the antibody binds CSF1R. In some embodiments, a humanized anti-CSFIR antibody comprises a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the antibody binds CSF1R. In some embodiments, a humanized anti-CSFIR antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45; and a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52; wherein the antibody binds CSF1R.

[0084] As used herein, whether a particular polypeptide is, for example, at least 95% identical to an amino acid sequence can be determined using, e.g., a computer program. When determining whether a particular sequence is, for example, 95% identical to a reference sequence, the percentage of identity is calculated over the full length of the reference amino acid sequence.

[0085] In some embodiments, a humanized anti-CSFIR antibody comprises at least one of the CDRs discussed herein. That is, in some embodiments, a humanized anti-CSFIR antibody comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, a heavy chain CDR3 discussed herein, a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, a humanized anti-CSFIR antibody comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the antibody comprising the mutated CDR.

[0086] Exemplary humanized anti-CSFIR antibodies also include antibodies that compete for binding to CSF1R with an antibody described herein. Thus, in some embodiments, a humanized anti-CSFIR antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311; and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.

Exemplary humanized antibody constant regions



[0087] In some embodiments, a humanized antibody described herein comprises one or more human constant regions. In some embodiments, the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD. In some embodiments, the human light chain constant region is of an isotype selected from κ and λ. In some embodiments, a humanized antibody described herein comprises a human IgG constant region. In some embodiments, a humanized antibody described herein comprises a human IgG4 heavy chain constant region. In some such embodiments, a humanized antibody described herein comprises an S241P mutation in the human IgG4 constant region. In some embodiments, a humanized antibody described herein comprises a human IgG4 constant region and a human κ light chain.

[0088] The choice of heavy chain constant region can determine whether or not an antibody will have effector function in vivo. Such effector function, in some embodiments, includes antibody-dependent cell-mediated cytotoxicity (ADCC) and/or complement-dependent cytotoxicity (CDC), and can result in killing of the cell to which the antibody is bound. In some methods of treatment, including methods of treating some cancers, cell killing may be desirable, for example, when the antibody binds to a cell that supports the maintenance or growth of the tumor. Exemplary cells that may support the maintenance or growth of a tumor include, but are not limited to, tumor cells themselves, cells that aid in the recruitment of vasculature to the tumor, and cells that provide ligands, growth factors, or counter-receptors that support or promote tumor growth or tumor survival. In some embodiments, when effector function is desirable, an anti-CSFIR antibody comprising a human IgG1 heavy chain or a human IgG3 heavy chain is selected.

[0089] An antibody may be humanized by any method. Nonlimiting exemplary methods of humanization include methods described, e.g., in U.S. Patent Nos. 5,530,101; 5,585,089; 5,693,761; 5,693,762; 6,180,370; Jones et al., Nature 321: 522-525 (1986); Riechmann et al., Nature 332: 323-27 (1988); Verhoeyen et al., Science 239: 1534-36 (1988); and U.S. Publication No. US 2009/0136500.

[0090] As noted above, a humanized antibody is an antibody in which at least one amino acid in a framework region of a non-human variable region has been replaced with the amino acid from the corresponding location in a human framework region. In some embodiments, at least two, at least three, at least four, at least five, at least six, at least seven, at least eight, at least nine, at least 10, at least 11, at least 12, at least 15, or at least 20 amino acids in the framework regions of a non-human variable region are replaced with an amino acid from one or more corresponding locations in one or more human framework regions.

[0091] In some embodiments, some of the corresponding human amino acids used for substitution are from the framework regions of different human immunoglobulin genes. That is, in some such embodiments, one or more of the non-human amino acids may be replaced with corresponding amino acids from a human framework region of a first human antibody or encoded by a first human immunoglobulin gene, one or more of the non-human amino acids may be replaced with corresponding amino acids from a human framework region of a second human antibody or encoded by a second human immunoglobulin gene, one or more of the non-human amino acids may be replaced with corresponding amino acids from a human framework region of a third human antibody or encoded by a third human immunoglobulin gene, etc. Further, in some embodiments, all of the corresponding human amino acids being used for substitution in a single framework region, for example, FR2, need not be from the same human framework. In some embodiments, however, all of the corresponding human amino acids being used for substitution are from the same human antibody or encoded by the same human immunoglobulin gene.

[0092] In some embodiments, an antibody is humanized by replacing one or more entire framework regions with corresponding human framework regions. In some embodiments, a human framework region is selected that has the highest level of homology to the non-human framework region being replaced. In some embodiments, such a humanized antibody is a CDR-grafted antibody.

[0093] In some embodiments, following CDR-grafting, one or more framework amino acids are changed back to the corresponding amino acid in a mouse framework region. Such "back mutations" are made, in some embodiments, to retain one or more mouse framework amino acids that appear to contribute to the structure of one or more of the CDRs and/or that may be involved in antigen contacts and/or appear to be involved in the overall structural integrity of the antibody. In some embodiments, ten or fewer, nine or fewer, eight or fewer, seven or fewer, six or fewer, five or fewer, four or fewer, three or fewer, two or fewer, one, or zero back mutations are made to the framework regions of an antibody following CDR grafting.

[0094] In some embodiments, a humanized antibody also comprises a human heavy chain constant region and/or a human light chain constant region.

Exemplary Chimeric Antibodies



[0095] In some embodiments, an anti-CSFIR antibody is a chimeric antibody. In some embodiments, an anti-CSFIR antibody comprises at least one non-human variable region and at least one human constant region. In some such embodiments, all of the variable regions of an anti-CSFIR antibody are non-human variable regions, and all of the constant regions of an anti-CSFIR antibody are human constant regions. In some embodiments, one or more variable regions of a chimeric antibody are mouse variable regions. The human constant region of a chimeric antibody need not be of the same isotype as the non-human constant region, if any, it replaces. Chimeric antibodies are discussed, e.g., in U.S. Patent No. 4,816,567; and Morrison et al. Proc. Natl. Acad. Sci. USA 81: 6851-55 (1984).

[0096] Nonlimiting exemplary chimeric antibodies include chimeric antibodies comprising the heavy and/or light chain variable regions of an antibody selected from 0301, 0302, and 0311. Additional nonlimiting exemplary chimeric antibodies include chimeric antibodies comprising heavy chain CDR1, CDR2, and CDR3, and/or light chain CDR1, CDR2, and CDR3 of an antibody selected from 0301, 0302, and 0311.

[0097] Nonlimiting exemplary chimeric anti-CSFIR antibodies include antibodies comprising the following pairs of heavy and light chain variable regions: SEQ ID NOs: 9 and 10; SEQ ID NOs: 11 and 12; and SEQ ID NOs: 13 and 14.

[0098] Nonlimiting exemplary anti-CSFIR antibodies include antibodies comprising a set of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 shown above in Table 1.

Further exemplary chimeric antibodies



[0099] In some embodiments, a chimeric anti-CSFIR antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, wherein the antibody binds CSF1R. In some embodiments, a chimeric anti-CSFIR antibody comprises a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the antibody binds CSF1R. In some embodiments, a chimeric anti-CSFIR antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45; and a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52; wherein the antibody binds CSF1R.

[0100] In some embodiments, a chimeric anti-CSFIR antibody comprises at least one of the CDRs discussed herein. That is, in some embodiments, a chimeric anti-CSFIR antibody comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, a heavy chain CDR3 discussed herein, a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, a chimeric anti-CSFIR antibody comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the antibody comprising the mutated CDR.

[0101] Exemplary chimeric anti-CSFIR antibodies also include chimeric antibodies that compete for binding to CSF1R with an antibody described herein. Thus, in some embodiments, a chimeric anti-CSFIR antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311; and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.

Exemplary chimeric antibody constant regions



[0102] In some embodiments, a chimeric antibody described herein comprises one or more human constant regions. In some embodiments, the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD. In some embodiments, the human light chain constant region is of an isotype selected from κ and λ. In some embodiments, a chimeric antibody described herein comprises a human IgG constant region. In some embodiments, a chimeric antibody described herein comprises a human IgG4 heavy chain constant region. In some such embodiments, a chimeric antibody described herein comprises an S241P mutation in the human IgG4 constant region. In some embodiments, a chimeric antibody described herein comprises a human IgG4 constant region and a human κ light chain.

[0103] As noted above, whether or not effector function is desirable may depend on the particular method of treatment intended for an antibody. Thus, in some embodiments, when effector function is desirable, a chimeric anti-CSFIR antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is not desirable, a chimeric anti-CSFIR antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.

Exemplary Human Antibodies



[0104] Human antibodies can be made by any suitable method. Nonlimiting exemplary methods include making human antibodies in transgenic mice that comprise human immunoglobulin loci. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA 90: 2551-55 (1993); Jakobovits et al., Nature 362: 255-8 (1993); Lonberg et al., Nature 368: 856-9 (1994); and U.S. Patent Nos. 5,545,807; 6,713,610; 6,673,986; 6,162,963; 5,545,807; 6,300,129; 6,255,458; 5,877,397; 5,874,299; and 5,545,806.

[0105] Nonlimiting exemplary methods also include making human antibodies using phage display libraries. See, e.g., Hoogenboom et al., J. Mol. Biol. 227: 381-8 (1992); Marks et al., J. Mol. Biol. 222: 581-97 (1991); and PCT Publication No. WO 99/10494.

[0106] In some embodiments, a human anti-CSFIR antibody binds to a polypeptide having the sequence of SEQ ID NO: 1. Exemplary human anti-CSFIR antibodies also include antibodies that compete for binding to CSF1R with an antibody described herein. Thus, in some embodiments, a human anti-CSFIR antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311, and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.

[0107] In some embodiments, a human anti-CSFIR antibody comprises one or more human constant regions. In some embodiments, the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD. In some embodiments, the human light chain constant region is of an isotype selected from κ and λ. In some embodiments, a human antibody described herein comprises a human IgG constant region. In some embodiments, a human antibody described herein comprises a human IgG4 heavy chain constant region. In some such embodiments, a human antibody described herein comprises an S241P mutation in the human IgG4 constant region. In some embodiments, a human antibody described herein comprises a human IgG4 constant region and a human κ light chain.

[0108] In some embodiments, when effector function is desirable, a human anti-CSFIR antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is not desirable, a human anti-CSFIR antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.

Additional Exemplary Anti-CSFIR Antibodies



[0109] Exemplary anti-CSFIR antibodies also include, but are not limited to, mouse, humanized, human, chimeric, and engineered antibodies that comprise, for example, one or more of the CDR sequences described herein. In some embodiments, an anti-CSFIR antibody comprises a heavy chain variable region described herein. In some embodiments, an anti-CSF1R antibody comprises a light chain variable region described herein. In some embodiments, an anti-CSFIR antibody comprises a heavy chain variable region described herein and a light chain variable region described herein. In some embodiments, an anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3 described herein. In some embodiments, an anti-CSFIR antibody comprises light chain CDR1, CDR2, and CDR3 described herein. In some embodiments, an anti-CSF1R antibody comprises heavy chain CDR1, CDR2, and CDR3 described herein and light chain CDR1, CDR2, and CDR3 described herein.

[0110] In some embodiments, an anti-CSFIR antibody comprises a heavy chain variable region of an antibody selected from Fabs 0301, 0302, and 0311. Nonlimiting exemplary anti-CSFIR antibodies also include antibodies comprising a heavy chain variable region of an antibody selected from humanized antibodies huAb1 to huAb16. Nonlimiting exemplary anti-CSFIR antibodies include antibodies comprising a heavy chain variable region comprising a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45.

[0111] In some embodiments, an anti-CSFIR antibody comprises a light chain variable region of an antibody selected from Fabs 0301, 0302, and 0311. Nonlimiting exemplary anti-CSF1R antibodies also include antibodies comprising a light chain variable region of an antibody selected from humanized antibodies huAb1 to huAb16. Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising a light chain variable region comprising a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52.

[0112] In some embodiments, an anti-CSFIR antibody comprises a heavy chain variable region and a light chain variable region of an antibody selected from Fabs 0301, 0302, and 0311. Nonlimiting exemplary anti-CSFIR antibodies also include antibodies comprising a heavy chain variable region and a light chain variable region of an antibody selected from humanized antibodies huAb1 to huAb16. Nonlimiting exemplary anti-CSFIR antibodies include antibodies comprising the following pairs of heavy and light chain variable regions: SEQ ID NOs: 9 and 10; SEQ ID NOs: 11 and 12; and SEQ ID NOs: 13 and 14; SEQ ID NOs: 39 and 40; SEQ ID NOs: 41 and 42; SEQ ID NOs: 43 and 44; SEQ ID NOs: 45 and 46; SEQ ID NOs: 47 and 48; SEQ ID NOs: 49 and 50; and SEQ ID NOs: 51 and 52. Nonlimiting exemplary anti-CSFIR antibodies also include antibodies comprising the following pairs of heavy and light chains: SEQ ID NOs: 33 and 34; SEQ ID NOs: 35 and 36; and SEQ ID NOs: 37 and 38.

[0113] In some embodiments, an anti-CSFIR antibody comprises heavy chain CDR1, CDR2, and CDR3 of an antibody selected from Fabs 0301, 0302, and 0311. Nonlimiting exemplary anti-CSFIR antibodies include antibodies comprising sets of heavy chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 15, 16, and 17; SEQ ID NOs: 21, 22, and 23; and SEQ ID NOs: 27, 28,and 29.

[0114] In some embodiments, an anti-CSFIR antibody comprises light chain CDR1, CDR2, and CDR3 of an antibody selected from Fabs 0301, 0302, and 0311. Nonlimiting exemplary anti-CSF1R antibodies include antibodies comprising sets of light chain CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 18, 19, and 20; SEQ ID NOs: 24, 25, and 26; and SEQ ID NOs: 30, 31, and 32.

[0115] In some embodiments, an anti-CSFIR antibody comprises heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 of an antibody selected from Fabs 0301, 0302, and 0311.

[0116] Nonlimiting exemplary anti-CSFIR antibodies include antibodies comprising the sets of heavy chain CDR1, CDR2, and CDR3, and light chain CDR1, CDR2, and CDR3 shown above in Table 1.

Further exemplary antibodies



[0117] In some embodiments, an anti-CSFIR antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, wherein the antibody binds CSF1R. In some embodiments, an anti-CSFIR antibody comprises a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the antibody binds CSF1R. In some embodiments, an anti-CSFIR antibody comprises a heavy chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45; and a light chain comprising a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52; wherein the antibody binds CSF1R.

[0118] In some embodiments, an anti-CSFIR antibody comprises at least one of the CDRs discussed herein. That is, in some embodiments, an anti-CSFIR antibody comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, a heavy chain CDR3 discussed herein, a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, an anti-CSFIR antibody comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the antibody comprising the mutated CDR.

[0119] Exemplary anti-CSFIR antibodies also include antibodies that compete for binding to CSF1R with an antibody described herein. Thus, in some embodiments, an anti-CSF1R antibody is provided that competes for binding to CSF1R with an antibody selected from Fabs 0301, 0302, and 0311, and bivalent (i.e., having two heavy chains and two light chains) antibody versions of those Fabs.

Exemplary antibody constant regions



[0120] In some embodiments, an antibody described herein comprises one or more human constant regions. In some embodiments, the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD. In some embodiments, the human light chain constant region is of an isotype selected from κ and λ. In some embodiments, an antibody described herein comprises a human IgG constant region. In some embodiments, an antibody described herein comprises a human IgG4 heavy chain constant region. In some such embodiments, an antibody described herein comprises an S241P mutation in the human IgG4 constant region. In some embodiments, an antibody described herein comprises a human IgG4 constant region and a human κ light chain.

[0121] As noted above, whether or not effector function is desirable may depend on the particular method of treatment intended for an antibody. Thus, in some embodiments, when effector function is desirable, an anti-CSFIR antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is not desirable, an anti-CSFIR antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.

Exemplary Anti-CSFIR Heavy Chain Variable Regions



[0122] In some embodiments, anti-CSFIR antibody heavy chain variable regions are provided. In some embodiments, an anti-CSFIR antibody heavy chain variable region is a mouse variable region, a human variable region, or a humanized variable region.

[0123] An anti-CSFIR antibody heavy chain variable region comprises a heavy chain CDR1, FR2, CDR2, FR3, and CDR3. In some embodiments, an anti-CSFIR antibody heavy chain variable region further comprises a heavy chain FR1 and/or FR4. Nonlimiting exemplary heavy chain variable regions include, but are not limited to, heavy chain variable regions having an amino acid sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45.

[0124] In some embodiments, an anti-CSFIR antibody heavy chain variable region comprises a CDR1 comprising a sequence selected from SEQ ID NOs: 15, 21, and 27.

[0125] In some embodiments, an anti-CSFIR antibody heavy chain variable region comprises a CDR2 comprising a sequence selected from SEQ ID NOs: 16, 22, and 28.

[0126] In some embodiments, an anti-CSFIR antibody heavy chain variable region comprises a CDR3 comprising a sequence selected from SEQ ID NOs: 17, 23, and 29.

[0127] Nonlimiting exemplary heavy chain variable regions include, but are not limited to, heavy chain variable regions comprising sets of CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 15, 16, and 17; SEQ ID NOs: 21, 22, and 23; and SEQ ID NOs: 27, 28, and 29.

[0128] In some embodiments, an anti-CSFIR antibody heavy chain comprises a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 9, 11, 13, and 39 to 45, wherein the heavy chain, together with a light chain, is capable of forming an antibody that binds CSF1R.

[0129] In some embodiments, an anti-CSFIR antibody heavy chain comprises at least one of the CDRs discussed herein. That is, in some embodiments, an anti-CSFIR antibody heavy chain comprises at least one CDR selected from a heavy chain CDR1 discussed herein, a heavy chain CDR2 discussed herein, and a heavy chain CDR3 discussed herein. Further, in some embodiments, an anti-CSFIR antibody heavy chain comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the heavy chain comprising the mutated CDR.

[0130] In some embodiments, a heavy chain comprises a heavy chain constant region. In some embodiments, a heavy chain comprises a human heavy chain constant region. In some embodiments, the human heavy chain constant region is of an isotype selected from IgA, IgG, and IgD. In some embodiments, the human heavy chain constant region is an IgG constant region. In some embodiments, a heavy chain comprises a human igG4 heavy chain constant region. In some such embodiments, the human IgG4 heavy chain constant region comprises an S241P mutation.

[0131] In some embodiments, when effector function is desirable, a heavy chain comprises a human IgG1 or IgG3 heavy chain constant region. In some embodiments, when effector function is less desirable, a heavy chain comprises a human IgG4 or IgG2 heavy chain constant region.

Exemplary Anti-CSFIR Light Chain Variable Regions



[0132] In some embodiments, anti-CSFIR antibody light chain variable regions are provided. In some embodiments, an anti-CSFIR antibody light chain variable region is a mouse variable region, a human variable region, or a humanized variable region.

[0133] An anti-CSFIR antibody light chain variable region comprises a light chain CDR1, FR2, CDR2, FR3, and CDR3. In some embodiments, an anti-CSFIR antibody light chain variable region further comprises a light chain FR1 and/or FR4. Nonlimiting exemplary light chain variable regions include light chain variable regions having an amino acid sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52.

[0134] In some embodiments, an anti-CSFIR antibody light chain variable region comprises a CDR1 comprising a sequence selected from SEQ ID NOs: 18, 24 and 30.

[0135] In some embodiments, an anti-CSFIR antibody light chain variable region comprises a CDR2 comprising a sequence selected from SEQ ID NOs: 19, 25, and 31.

[0136] In some embodiments, an anti-CSFIR antibody light chain variable region comprises a CDR3 comprising a sequence selected from SEQ ID NOs: 20, 26, and 32.

[0137] Nonlimiting exemplary light chain variable regions include, but are not limited to, light chain variable regions comprising sets of CDR1, CDR2, and CDR3 selected from: SEQ ID NOs: 18, 19, and 20; SEQ ID NOs: 24, 25, and 26; and SEQ ID NOs: 30, 31, and 32.

[0138] In some embodiments, an anti-CSFIR antibody light chain comprises a variable region sequence that is at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% identical to a sequence selected from SEQ ID NOs: 10, 12, 14, and 46 to 52, wherein the light chain, together with a heavy chain, is capable of forming an antibody that binds CSF1R.

[0139] In some embodiments, an anti-CSFIR antibody light chain comprises at least one of the CDRs discussed herein. That is, in some embodiments, an anti-CSFIR antibody light chain comprises at least one CDR selected from a light chain CDR1 discussed herein, a light chain CDR2 discussed herein, and a light chain CDR3 discussed herein. Further, in some embodiments, an anti-CSFIR antibody light chain comprises at least one mutated CDR based on a CDR discussed herein, wherein the mutated CDR comprises 1, 2, 3, or 4 amino acid substitutions relative to the CDR discussed herein. In some embodiments, one or more of the amino acid substitutions are conservative amino acid substitutions. One skilled in the art can select one or more suitable conservative amino acid substitutions for a particular CDR sequence, wherein the suitable conservative amino acid substitutions are not predicted to significantly alter the binding properties of the light chain comprising the mutated CDR.

[0140] In some embodiments, a light chain comprises a human light chain constant region. In some embodiments, a human light chain constant region is selected from a human κ and a human λ light chain constant region.

Exemplary Additional CSF1R Binding Molecules



[0141] In some embodiments, additional molecules that bind CSF1R are provided. Such molecules include, but are not limited to, non-canonical scaffolds, such as anti-calins, adnectins, ankyrin repeats, etc. See, e.g., Hosse et al., Prot. Sci. 15:14 (2006); Fiedler, M. and Skerra, A., "Non-Antibody Scaffolds," pp.467-499 in Handbook of Therapeutic Antibodies, Dubel, S., ed., Wiley-VCH, Weinheim, Germany, 2007.

Exemplary Properties of anti-CSF 1R antibodies



[0142] In some embodiments, an antibody having a structure described above binds to the CSF1R with a binding affinity (KD) of less than 1 nM, blocks binding of CSF1 and/or IL-34 to CSF1R, and inhibits CSF1R phosphorylation induced by CSF1 and/or IL-34.

[0143] In some embodiments, an anti-CSFIR antibody binds to the extracellular domain of CSF1R (CSF1R-ECD). In some embodiments, an anti-CSFIR antibody has a binding affinity (KD) for CSF1R of less than 1 nM, less than 0.5 nM, less than 0.1 nM, or less than 0.05 nM. In some embodiments, an anti-CSFIR antibody has a KD of between 0.01 and 1 nM, between 0.01 and 0.5 nM, between 0.01 and 0.1 nM, between 0.01 and 0.05 nM, or between 0.02 and 0.05 nM.

[0144] In some embodiments, an anti-CSFIR antibody blocks ligand binding to CSF1R. In some embodiments, an anti-CSFIR antibody blocks binding of CSF1 to CSF1R. In some embodiments, an anti-CSFIR antibody blocks binding of IL-34 to CSF1R. In some embodiments, an anti-CSFIR antibody blocks binding of both CSF1 and IL-34 to CSF1R. In some embodiments, an antibody that blocks ligand binding binds to the extracellular domain of CSF1R. In some embodiments, an antibody blocks ligand binding to CSF1R when it reduces the amount of detectable binding of a ligand to CSF1R by at least 50%, using the assay described, e.g., U.S. Patent No. 8,206,715 B2, Example 7. In some embodiments, an antibody reduces the amount of detectable binding of a ligand to CSF1R by at least 60%, at least 70%, at least 80%, or at least 90%. In some such embodiments, the antibody is said to block ligand binding by at least 50%, at least 60%, at least 70%, etc.

[0145] In some embodiments, an anti-CSFIR antibody inhibits ligand-induced CSF1R phosphorylation. In some embodiments, an anti-CSF1R antibody inhibits CSF1-induced CSF1R phosphorylation. In some embodiments, an anti-CSFIR antibody inhibits IL-34-induced CSF1R phosphorylation. In some embodiments, an anti-CSFIR antibody inhibits both CSF1-induced and IL-34-induced CSF1R phosphorylation. In some embodiments, an antibody is considered to "inhibit ligand-induced CSF1R phosphorylation" when it reduces the amount of detectable ligand-induced CSF1R phosphorylation by at least 50%, using the assay described, e.g., U.S. Patent No. 8,206,715 B2, Example 6. In some embodiments, an antibody reduces the amount of detectable ligand-induced CSF1R phosphorylation by at least 60%, at least 70%, at least 80%, or at least 90%. In some such embodiments, the antibody is said to inhibit ligand-induced CSF1R phosphorylation by at least at least 50%, at least 60%, at least 70%, etc.

[0146] In some embodiments, an antibody inhibits monocyte proliferation and/or survival responses in the presence of CSF1 and/or IL-34. In some embodiments, an antibody is considered to "inhibit monocyte proliferation and/or survival responses" when it reduces the amount of monocyte proliferation and/or survival responses in the presence of CSF1 and/or IL-34 by at least 50%, using the assay described, e.g., U.S. Patent No. 8,206,715 B2, Example 10. In some embodiments, an antibody reduces the amount of monocyte proliferation and/or survival responses in the presence of CSF1 and/or IL-34 by at least 60%, at least 70%, at least 80%, or at least 90%. In some such embodiments, the antibody is said to inhibit monocyte proliferation and/or survival responses by at least at least 50%, at least 60%, at least 70%, etc.

Exemplary Immune Stimulating Agents



[0147] The immune stimulating agents that are included in the compositions for use according to the invention (or with which the compositions for use according to the invention are to be used) are as defined in the claims. The remaining disclosure of this section is subject to the requirements of the claims.

[0148] Immune stimulating agents may include, for example, a small molecule drug, antibody or fragment thereof, or other biologic or small molecule. Examples of biologic immune stimulating agents include, but are not limited to, antibodies, antibody fragments, fragments of receptor or ligand polypeptides, for example that block receptor-ligand binding, vaccines and cytokines. In one aspect, the antibody is a monoclonal antibody. In certain aspects, the monoclonal antibody is humanized or human antibody.

[0149] In some embodiments, the at least one immune stimulating agent comprises an agonist of an immune stimulatory molecule, including a co-stimulatory molecule, while in some embodiments, the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule. In some embodiments, the at least one immune stimulating agent comprises an agonist of an immune-stimulatory molecule, including a co-stimulatory molecule, found on immune cells, such as T cells. In some embodiments, the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule, found on immune cells, such as T cells. In some embodiments, the at least one immune stimulating agent comprises an agonist of an immune stimulatory molecule, including a co-stimulatory molecule, found on cells involved in innate immunity, such as NK cells. In some embodiments, the at least one immune stimulating agent comprises an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule, found on cells involved in innate immunity, such as NK cells. In some embodiments, the combination enhances the antigen-specific T cell response in the treated subject and/or enhances the innate immunity response in the subject. In some embodiments, the combination results in an improved anti-tumor response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSFIR antibody or immune stimulating agent alone. In some embodiments, the combination results in a synergistic response in an animal cancer model, such as a xenograft model, compared to administration of either the anti-CSFIR antibody or immune stimulating agent alone.

[0150] In certain embodiments, an immune stimulating agent targets a stimulatory or inhibitory molecule that is a member of the immunoglobulin super family (IgSF). For example, an immune stimulating agent may be an agent that targets (or binds specifically to) a member of the B7 family of membrane-bound ligands, which includes B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6, or a co-stimulatory or co-inhibitory receptor binding specifically to a B7 family member. An immune stimulating agent may be an agent that targets a member of the TNF family of membrane bound ligands or a co-stimulatory or co-inhibitory receptor binding specifically to a member of the TNF family. Exemplary TNF and TNFR family members that may be targeted by immune stimulating agents include CD40 and CD40L, OX-40, OX-40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, TACI, APRIL, BCMA, LTβR, LIGHT, DcR3, HVEM, VEGI/TL1A, TRAMP/DR3, EDAR, EDA1, XEDAR, EDA2, TNFR1, Lymphotoxin α/TNEβ, TNFR2, TNFα, LTβR, Lymphotoxin α 1β2, FAS, FASL, RELT, DR6, TROY and NGFR. An immune stimulating agent may be an agent, e.g., an antibody, targeting an IgSF member, such as a B7 family member, a B7 receptor family member, a TNF family member or a TNFR family member, such as those described above.

[0151] In some embodiments, an immune stimulating agent may comprise (i) an antagonist of a protein that inhibits T cell activation (e.g., immune checkpoint inhibitor) such as CTLA-4, LAG-3, TIM3, Galectin 9, CEACAM-1, BTLA, CD69, Galectin-1, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM-1, TIM-4, and ILT4 and/or may comprise (ii)an agonist of a protein that stimulates T cell activation such as B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD70, CD27, CD40, CD40L, DR3 and CD28H.

[0152] In some embodiments, an immune stimulating agent may comprise an agent that inhibit or is an antagonist of a cytokine that inhibits T cell activation (e.g., IL-6, IL-10, TGF-β, VEGF, and other immunosuppressive cytokines), and it some embodiments an immune stimulating agent may comprise an agent that is an agonist of a cytokine, such as IL-2, IL-7, IL-12, IL-15, IL-21 and IFNα (e.g., the cytokine itself) that stimulates T cell activation. In some embodiments, immune stimulating agents may comprise an antagonist of a chemokine, such as CXCR2 (e.g., MK-7123), CXCR4 (e.g. AMD3100), CCR2, or CCR4 (mogamulizumab).

[0153] In some embodiments, immune stimulating agents may include antagonists of inhibitory receptors on NK cells or agonists of activating receptors on NK cells. For example, an anti-CSFIR antibody can be combined with an antagonist of KIR, optionally along with at least one other immune stimulating agent such as an agonist of CD40.

[0154] Immune stimulating agents may also include agents that inhibit TGF-β signaling, agents that enhance tumor antigen presentation, e.g., dendritic cell vaccines, GM-CSF secreting cellular vaccines, CpG oligonucleotides,and imiquimod, or therapies that enhance the immunogenicity of tumor cells (e.g., anthracyclines).

[0155] Immune stimulating agents may also include certain vaccines such as mesothelin-targeting vaccines or attenuated listeria cancer vaccines, such as CRS-207.

[0156] Immune stimulating agents may also comprise agents that deplete or block Treg cells, such as agents that specifically bind to CD25.

[0157] Immune stimulating agents may also comprise agents that inhibit a metabolic enzyme such as indoleamine dioxigenase (IDO), dioxigenase, arginase, or nitric oxide synthetase.

[0158] Immune stimulating agents may also comprise agents that inhibit the formation of adenosine or inhibit the adenosine A2A receptor.

[0159] Immune stimulating agents may also comprise agents that reverse/prevent T cell anergy or exhaustion and agents that trigger an innate immune activation and/or inflammation at a tumor site.

[0160] An anti-CSFIR antibody may be combined with more than one immune stimulating agent, such as a CD40 agonist and at least one additional immune stimulating agent. The anti-CSFIR antibody, optionally along with a CD40 agonist, may be combined with a combinatorial approach that targets multiple elements of the immune pathway, such as one or more of the following: at least one agent that enhances tumor antigen presentation (e.g., dendritic cell vaccine, GM-CSF secreting cellular vaccines, CpG oligonucleotides, imiquimod); at least one agent that inhibits negative immune regulation e.g., by inhibiting CTLA-4 pathway and/or depleting or blocking Treg or other immune suppressing cells; a therapy that stimulates positive immune regulation, e.g., with agonists that stimulate the CD-137, OX-40 and/or GITR pathway and/or stimulate T cell effector function; at least one agent that increases systemically the frequency of anti-tumor T cells; a therapy that depletes or inhibits Tregs, such as Tregs in the tumor, e.g., using an antagonist of CD25 (e.g., daclizumab) or by ex vivo anti-CD25 bead depletion; at least one agent that impacts the function of suppressor myeloid cells in the tumor; a therapy that enhances immunogenicity of tumor cells (e.g., anthracyclines); adoptive T cell or NK cell transfer including genetically modified cells, e.g., cells modified by chimeric antigen receptors (CAR-T therapy); at least one agent that inhibits a metabolic enzyme such as indoleamine dioxigenase (IDO), dioxigenase, arginase or nitric oxide synthetase; at least one agent that reverses/prevents T cell anergy or exhaustion; a therapy that triggers an innate immune activation and/or inflammation at a tumor site; administration of immune stimulatory cytokines or blocking of immuno repressive cytokines.

[0161] For example, an anti-CSFIR antibody, optionally with a CD40 agonist, can be used with one or more agonistic agents that ligate positive costimulatory receptors; one or more antagonists (blocking agents) that attenuate signaling through inhibitory receptors, such as antagonists that overcome distinct immune suppressive pathways within the tumor microenvironment; one or more agents that increase systemically the frequency of anti-tumor immune cells, such as T cells, deplete or inhibit Tregs (e.g., by inhibiting CD25); one or more agents that inhibit metabolic enzymes such as IDO; one or more agents that reverse/prevent T cell anergy or exhaustion; and one or more agents that trigger innate immune activation and/or inflammation at tumor sites.

[0162] In one embodiment, the at least one immune stimulating agent comprises a CTLA-4 antagonist, such as an antagonistic CTLA-4 antibody. Suitable CTLA-4 antibodies include, for example, YERVOY (ipilimumab) or tremelimumab.

[0163] In some embodiments, the at least one immune stimulating agent comprises a LAG-3 antagonist, such as an antagonistic LAG-3 antibody. Suitable LAG3 antibodies include, for example, BMS-986016 (WO10/19570, WO14/08218), or IMP-731 or IMP-321 (WO08/132601, WO09/44273).

[0164] In some embodiments, the at least one immune stimulating agent comprises a CD137 (4-1BB) agonist, such as an agonistic CD137 antibody. Suitable CD137 antibodies include, for example, urelumab or PF-05082566 (WO12/32433).

[0165] In some embodiments, the at least one immune stimulating agent comprises a GITR agonist, such as an agonistic GITR antibody. Suitable GITR antibodies include, for example, TRX-518 (WO06/105021, WO09/009116), MK-4166 (WO11/028683) or a GITR antibody disclosed in WO2015/031667.

[0166] In some embodiments, the at least one immune stimulating agent comprises an OX40 agonist, such as an agonistic OX40 antibody. Suitable OX40 antibodies include, for example, MEDI-6383, MEDI-6469 or MOXR0916 (RG7888; WO06/029879).

[0167] In some embodiments, the at least one immune stimulating agent comprises a CD27 agonist, such as an agonistic CD27 antibody. Suitable CD27 antibodies include, for example, varlilumab (CDX-1127).

[0168] In some embodiments, the at least one immune stimulating agent comprises MGA271, which targets B7H3 (WO11/109400).

[0169] In some embodiments, the at least one immune stimulating agent comprises a KIR antagonist, such as lirilumab.

[0170] In some embodiments, the at least one immune stimulating agent comprises an IDO antagonist. IDO antagonists include, for example, INCB-024360 (WO2006/122150, WO07/75598, WO08/36653, WO08/36642), indoximod, NLG-919 (WO09/73620, WO09/1156652, WO11/56652, WO12/142237) or F001287.

[0171] In some embodiments, the at least one immune stimulating agent comprises a Toll-like receptor agonist, e.g., a TLR2/4 agonist (e.g., Bacillus Calmette-Guerin); a TLR7 agonist (e.g., Hiltonol or Imiquimod); a TLR7/8 agonist (e.g., Resiquimod); or a TLR9 agonist (e.g., CpG7909).

[0172] In some embodiments, the at least one immune stimulating agent comprises a TGF-β inhibitor, e.g., GC1008, LY2157299, TEW7197 or IMC-TR1.

CD40 Agonists and Exemplary CD40 Agonist Molecules



[0173] In accordance with the invention, the at least one immune stimulating agent comprises an agonist anti-CD40 antibody, optionally together with at least one additional immune stimulating agent as described herein. The cell surface molecule CD40 is a member of the tumor necrosis factor receptor superfamily and is expressed by antigen presenting cells such as dentritic cells, B cells, macrophages and monocytes and is also expressed on other cell types, including immune, hematopoietic, vascular, and epithelial cells, as well as on various tumor cells. In antigen presenting cells CD40 signaling results in activation and upregulation of T cell costimulatory molecules and other critical immune mediators required for the induction of an immune response. Agonists of CD40 are potential cancer therapies, causing tumor regression through both anti-tumor immune activation and direct cytotoxic effect on tumor cells. CD40-targeting therapies have undergone phase 1 clinical evaluation in advanced-stage cancer patients, and initial findings have shown efficacy in the absence of major toxicity.

[0174] With regard to CD40, for example, animal models have shown that ligation of CD40 on dendritic cells results in activation of cytotoxic T lymphocytes that mediate tumor killing (Marzo et al., 2000, J. Immunol.; Todryk et al., 2001, J. Immunol. Methods.) Activation of CD40 on macrophages results in tumoralcidal activity (Beatty et al., 2011, Science), and cytokines produced from CD40 stimulated antigen presenting cells leads to the activation of natural killer cells important for tumor eradication. Given the complex nature of an anti-tumor immune response, effective cancer therapy may require combining multiple immunotherapy agents. Consistent with this, small molecule inhibition of CSF1R was shown to synergize with anti-PDl immune checkpoint blockade in a pancreatic tumor model. See Zhu et al., 2014, Cancer Res., 74: 5057-5069. Thus, tumors that have CSF1R-expressing TAMs may be sensitive to combination therapy with an anti-CSFIR antibody and a CD40 agonist.

[0175] Exemplary CD40 agonists of the compositions and methods of this invention include, for example, anti-CD40 antibodies that enhance CD40 activity. Such antibodies may be humanized antibodies, chimeric antibodies, mouse antibodies, human antibodies, and antibodies comprising the heavy chain and/or light chain CDRs of an anti-CD40 antibody iscussed herein.

[0176] Various agonist anti-CD40 antibodies are known in the art. Nonlimiting exemplary agonist anti-CD40 antibodies include, but are not limited to, CP-870,893 (Pfizer and VLST; antibody 21.4.1 in EP 1 476 185 B1 and US Patent No. 7,338,660; see also clinicaltrials.gov/ct2/show/NCT02225002); dacetuzumab (Seattle Genetics; SEQ ID NOs: 98 and 99 herein; see also US Patent No. 6,946,129 and US Patent No. 8,303,955); RO7009789 (Roche; see, e.g., clinicaltrials.gov/ct2/show/NCT02304393); ADC-1013 (Alligator Bioscience; US Publication No. 2014/0348836; see also clinicaltrials.gov/ct2/show/NCT02379741); SEA-CD40 (Seattle Genetics; afucosylated form of antibody comprising SEQ ID NOs: 98 and 99; see also clinicaltrials.gov/ct2/show/NCT02376699); and Chi Lob 7/4 (Univ. Southampton; US Publication No. 2009/0074711; see also clinicaltrials.gov/ct2/show/NCT01561911). See, e.g., Vonderheide et al., 2013, Clin Cancer Res 19:1035.

[0177] Exemplary CD40 agonists also include recombinant CD40L.

Exemplary Antibody Conjugates



[0178] In some embodiments, an antibody is conjugated to a label and/or a cytotoxic agent. As used herein, a label is a moiety that facilitates detection of the antibody and/or facilitates detection of a molecule to which the antibody binds. Nonlimiting exemplary labels include, but are not limited to, radioisotopes, fluorescent groups, enzymatic groups, chemiluminescent groups, biotin, epitope tags, metal-binding tags, etc. One skilled in the art can select a suitable label according to the intended application.

[0179] As used herein, a cytotoxic agent is a moiety that reduces the proliferative capacity of one or more cells. A cell has reduced proliferative capacity when the cell becomes less able to proliferate, for example, because the cell undergoes apoptosis or otherwise dies, the cell fails to proceed through the cell cycle and/or fails to divide, the cell differentiates, etc. Nonlimiting exemplary cytotoxic agents include, but are not limited to, radioisotopes, toxins, and chemotherapeutic agents. One skilled in the art can select a suitable cytotoxic according to the intended application.

[0180] In some embodiments, a label and/or a cytotoxic agent is conjugated to an antibody using chemical methods in vitro. Nonlimiting exemplary chemical methods of conjugation are known in the art, and include services, methods and/or reagents commercially available from, e.g., Thermo Scientific Life Science Research Produces (formerly Pierce; Rockford, IL), Prozyme (Hayward, CA), SACRI Antibody Services (Calgary, Canada), AbD Serotec (Raleigh, NC), etc. In some embodiments, when a label and/or cytotoxic agent is a polypeptide, the label and/or cytotoxic agent can be expressed from the same expression vector with at least one antibody chain to produce a polypeptide comprising the label and/or cytotoxic agent fused to an antibody chain. One skilled in the art can select a suitable method for conjugating a label and/or cytotoxic agent to an antibody according to the intended application.

Exemplary Leader Sequences



[0181] In order for some secreted proteins to express and secrete in large quantities, a leader sequence from a heterologous protein may be desirable. In some embodiments, a leader sequence is selected from SEQ ID NOs: 3 and 4, which are light chain and heavy chain leader sequences, respectively. In some embodiments, employing heterologous leader sequences may be advantageous in that a resulting mature polypeptide may remain unaltered as the leader sequence is removed in the ER during the secretion process. The addition of a heterologous leader sequence may be required to express and secrete some proteins.

[0182] Certain exemplary leader sequence sequences are described, e.g., in the online Leader sequence Database maintained by the Department of Biochemistry, National University of Singapore. See Choo et al., BMC Bioinformatics, 6: 249 (2005); and PCT Publication No. WO 2006/081430.

Nucleic Acid Molecules Encoding Antibodies



[0183] Nucleic acid molecules comprising polynucleotides that encode one or more chains of an antibody are provided. In some embodiments, a nucleic acid molecule comprises a polynucleotide that encodes a heavy chain or a light chain of an antibody. In some embodiments, a nucleic acid molecule comprises both a polynucleotide that encodes a heavy chain and a polynucleotide that encodes a light chain, of an antibody. In some embodiments, a first nucleic acid molecule comprises a first polynucleotide that encodes a heavy chain and a second nucleic acid molecule comprises a second polynucleotide that encodes a light chain.

[0184] In some such embodiments, the heavy chain and the light chain are expressed from one nucleic acid molecule, or from two separate nucleic acid molecules, as two separate polypeptides. In some embodiments, such as when an antibody is an scFv, a single polynucleotide encodes a single polypeptide comprising both a heavy chain and a light chain linked together.

[0185] In some embodiments, a polynucleotide encoding a heavy chain or light chain of an antibody comprises a nucleotide sequence that encodes a leader sequence, which, when translated, is located at the N terminus of the heavy chain or light chain. As discussed above, the leader sequence may be the native heavy or light chain leader sequence, or may be another heterologous leader sequence.

[0186] Nucleic acid molecules may be constructed using recombinant DNA techniques conventional in the art. In some embodiments, a nucleic acid molecule is an expression vector that is suitable for expression in a selected host cell.

Antibody Expression and Production


Vectors



[0187] Vectors comprising polynucleotides that encode antibody heavy chains and/or light chains are provided. Vectors comprising polynucleotides that encode antibody heavy chains and/or light chains are also provided. Such vectors include, but are not limited to, DNA vectors, phage vectors, viral vectors, retroviral vectors, etc. In some embodiments, a vector comprises a first polynucleotide sequence encoding a heavy chain and a second polynucleotide sequence encoding a light chain. In some embodiments, the heavy chain and light chain are expressed from the vector as two separate polypeptides. In some embodiments, the heavy chain and light chain are expressed as part of a single polypeptide, such as, for example, when the antibody is an scFv.

[0188] In some embodiments, a first vector comprises a polynucleotide that encodes a heavy chain and a second vector comprises a polynucleotide that encodes a light chain. In some embodiments, the first vector and second vector are transfected into host cells in similar amounts (such as similar molar amounts or similar mass amounts). In some embodiments, a mole- or mass-ratio of between 5:1 and 1:5 of the first vector and the second vector is transfected into host cells. In some embodiments, a mass ratio of between 1:1 and 1:5 for the vector encoding the heavy chain and the vector encoding the light chain is used. In some embodiments, a mass ratio of 1:2 for the vector encoding the heavy chain and the vector encoding the light chain is used.

[0189] In some embodiments, a vector is selected that is optimized for expression of polypeptides in CHO or CHO-derived cells, or in NSO cells. Exemplary such vectors are described, e.g., in Running Deer et al., Biotechnol. Prog. 20:880-889 (2004).

[0190] In some embodiments, a vector is chosen for in vivo expression of antibody heavy chains and/or antibody light chains in animals, including humans. In some such embodiments, expression of the polypeptide is under the control of a promoter that functions in a tissue-specific manner. For example, liver-specific promoters are described, e.g., in PCT Publication No. WO 2006/076288.

Host Cells



[0191] In various embodiments, antibody heavy chains and/or light chains may be expressed in prokaryotic cells, such as bacterial cells; or in eukaryotic cells, such as fungal cells (such as yeast), plant cells, insect cells, and mammalian cells. Such expression may be carried out, for example, according to procedures known in the art. Exemplary eukaryotic cells that may be used to express polypeptides include, but are not limited to, COS cells, including COS 7 cells; 293 cells, including 293-6E cells; CHO cells, including CHO-S and DG44 cells; PER.C6® cells (Crucell); and NSO cells. In some embodiments, antibody heavy chains and/or light chains may be expressed in yeast. See, e.g., U.S. Publication No. US 2006/0270045 A1. In some embodiments, a particular eukaryotic host cell is selected based on its ability to make desired post-translational modifications to the antibody heavy chains and/or light chains. For example, in some embodiments, CHO cells produce polypeptides that have a higher level of sialylation than the same polypeptide produced in 293 cells.

[0192] Introduction of one or more nucleic acids into a desired host cell may be accomplished by any method, including but not limited to, calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, etc. Nonlimiting exemplary methods are described, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual, 3rd ed. Cold Spring Harbor Laboratory Press (2001). Nucleic acids may be transiently or stably transfected in the desired host cells, according to any suitable method.

[0193] In some embodiments, one or more polypeptides may be produced in vivo in an animal that has been engineered or transfected with one or more nucleic acid molecules encoding the polypeptides, according to any suitable method.

Purification of Antibodies



[0194] Antibodies may be purified by any suitable method. Such methods include, but are not limited to, the use of affinity matrices or hydrophobic interaction chromatography. Suitable affinity ligands include the antigen and ligands that bind antibody constant regions. For example, a Protein A, Protein G, Protein A/G, or an antibody affinity column may be used to bind the constant region and to purify an antibody. Hydrophobic interactive chromatography, for example, a butyl or phenyl column, may also suitable for purifying some polypeptides. Many methods of purifying polypeptides are known in the art.

Cell-free Production of Antibodies



[0195] In some embodiments, an antibody is produced in a cell-free system. Nonlimiting exemplary cell-free systems are described, e.g., in Sitaraman et al., Methods Mol. Biol. 498: 229-44 (2009); Spirin, Trends Biotechnol. 22: 538-45 (2004); Endo et al., Biotechnol. Adv. 21: 695-713 (2003).

Therapeutic Compositions and Methods


Methods of Treating Cancer



[0196] In accordance with the invention as defined in the claims, compositions for use in methods for treating cancer are provided, the methods comprising administering an effective amount of an anti-CSF1R antibody and an effective amount of at least one immune stimulating agent. In some embodiments, the anti-CSFIR antibody and the at least one immune stimulating agent are administered concurrently. For example, the therapeutics may be infused together or injected at roughly the same time. In some embodiments, the anti-CSFIR antibody and the at least one immune stimulating agent administered sequentially. For example, in some embodiments the anti-CSFIR antibody is administered sequentially before or after at least one immune stimulating agent such that the two therapeutics are administered 30 minutes, 60 minutes, 90 minutes, 120 minutes, 3 hours, 6 hours, 12 hours, 24 hours, 36 hours, 48 hours, 3 days, 5 days, 7 days, or two weeks apart.

[0197] In some embodiments, at least one, at least two, at least three doses, at least five doses, or at least ten doses of an anti-CSF1R antibody is administered prior to administration of at least one immune stimulating agent. In some embodiments, at least one, at least two, at least three doses, at least five doses, or at least ten doses of at least one immune stimulating agent is administered prior to administration of an anti-CSFIR antibody. In some embodiments, the last dose of immune stimulating agent is administered at least one, two, three, five, days or ten, or one, two, three, five, twelve, or twenty four weeks prior to the first dose of CSFR1 inhibitor. In some other embodiment, the last dose of CSFR1 inhibitor is administered at least one, two, three, five, days or ten, or one, two, three, five, twelve, or twenty four weeks prior to the first dose of at least one immune stimulating agent. In some embodiments, a subject has received, or is receiving, therapy with at least one immune stimulating agent, and an anti-CSFIR antibody is added to the therapeutic regimen.

[0198] In some embodiments, a method of selecting a patient for combination therapy with an anti-CSFIR antibody and a at least one immune stimulating agent, such as a CD40 agonist is provided, comprising determining the levels of TAMs and/or CD8+ T cells in the patient. In some embodiments, if a patient's TAM levels are high, the patient is selected for combination therapy. In some embodiments, if a patient's TAM and CD8+ T cell levels are high, the patient is selected for combination therapy. The level of TAMs or CD8+ T cells is considered "high" if it is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, or at least 100% higher than the level in an individual who does not have cancer. In some embodiments, the level of TAMs or CD8+ T cells is considered "high" if it is above the median level found in individuals with cancer. In some embodiments, if a patient's TAM levels are high and CD8+ T cell levels are low, the patient is selected for combination therapy with an anti-CSFIR antibody and at least one immune stimulating agent, such as a CD40 agonist. The level of CD8+ T cells is considered "low" if it is at or below the median level found in individuals with cancer. In some embodiment, the level of CD8+ T cells is considered "low" if it is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 75%, or at least 100% lower than the level in an individual who does not have cancer. In some embodiments, expression of CSF1R on the patient's TAMs is determined. In some embodiments, if the patient's TAMs express CSF1R, the patient is selected for combination therapy. In some embodiments, if the patient's TAMs express elevated levels of CSF1R, the patient is selected for combination therapy. In some embodiments, a patient's TAMs are considered to express "elevated" levels of CSF1R if the level of CSF1R is at or above the median level of CSF1R found expressed on TAMS in individuals with cancer. In some embodiments, if the patient's CSF1R expression shows a high correlation with the level of CD8+ T cells, the patient is selected for combination therapy. The correlation of the expressions is considered "high" if it is at or above the median level found in individuals with cancer.

[0199] Levels of TAMs, CSF1R expression, CD8+ T cells, and/or regulatory T cells may be measured by methods in the art. Nonexemplary methods include immunohistochemistry (IHC), fluorescence-activated cell sorting (FACS), protein arrays, and gene expression assays, such as RNA sequencing, gene arrays, and quantitative PCR. In some embodiments, one or more markers selected from CSF1R, CD68, CD163, CD8, and FoxP3 may be detected by IHC, FACS, or gene expression assay on tumor sections, or dissociated cells from tumor sections.

[0200] In some embodiments, the cancer is selected from squamous cell cancer, small-cell lung cancer, pituitary cancer, esophageal cancer, astrocytoma, soft tissue sarcoma, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer, renal cancer, liver cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, brain cancer, endometrial cancer, testis cancer, cholangiocarcinoma, gallbladder carcinoma, gastric cancer, melanoma, and various types of head and neck cancer. In some embodiments, lung cancer is non-small cell lung cancer or lung squamous cell carcinoma. In some embodiments, leukemia is acute myeloid leukemia or chronic lymphocytic leukemia. In some embodiments, breast cancer is breast invasive carcinoma. In some embodiments, ovarian cancer is ovarian serous cystadenocarcinoma. In some embodiments, kidney cancer is kidney renal clear cell carcinoma. In some embodiments, colon cancer is colon adenocarcinoma. In some embodiments, bladder cancer is bladder urothelial carcinoma. In some embodiments, the cancer is selected from bladder cancer, cervical cancer (such as squamous cell cervical cancer), head and neck squamous cell carcinoma, rectal adenocarcinoma, non-small cell lung cancer, endometrial cancer, prostate adenocarcinoma, colon cancer, ovarian cancer (such as serous epithelial ovarian cancer), and melanoma.

[0201] In some embodiments, the anti-CSFIR antibody blocks binding of CSF1 and/or IL-34 to CSF1R and/or inhibits CSF1R phosphorylation induced by CSF1 and/or IL-34. In some embodiments, the anti-CSFIR antibody locks binding of CSF1 and IL-34 to CSF1R and/or inhibits CSF1R phosphorylation induced by CSF1 and/or IL-34. In some embodiments, the anti-CSFIR antibody comprises the CDRs of, or the variable regions of, an antibody selected from huAb1 to huAb16, described herein. In some embodiments, the anti-CSFIR antibody comprises the CDRs of, or the variable regions of, huAb1.

[0202] In some embodiments, the at least one immune stimulating agent comprises an antagonist of an inhibitor of the activation of T cells, while in some embodiments, the at least one immune stimulating agent comprises comprises an agonist of a stimulator of the activation of T cells. In some embodiments, the at least one immune stimulating agent comprises an antagonist of CTLA4, LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGFβ, VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO. In some embodiments, the at least one immune stimulating agent comprises an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFNα, STING, or a Toll-like receptor agonist such as a TLR2/4 agonist. In some embodiments, the at least one immune stimulating agent comprises an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6. In some embodiments, the at least one immune stimulating agent comprises an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LTβR, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNFβ, TNFR2, TNFα, 1β2, FAS, FASL, RELT, DR6, TROY, or NGFβ. In some embodiments, the at least one immune stimulating agent comprises an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGFβ, VEGF. In some embodiments, the at least one immune stimulating agent comprises an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFNα. In some embodiments, the at least one immune stimulating agent comprises an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4. In some embodiments, the at least one immune stimulating agent comprises an antibody. In some embodiments, the at least one immune stimulating agent may comprise a vaccine, such as a mesothelin-targeting vaccine or attenuated listeria cancer vaccine such as CRS-207.

[0203] In some embodiments, the at least one immune stimulating agent comprises a CD40 agonist, for example, an anti-CD40 antibody. Nonlimiting exemplary agonist anti-CD40 antibodies include CP-870,893 (Pfizer and VLST); dacetuzumab (Seattle Genetics); RO7009789 (Roche); ACD-1013 (Alligator Bioscience); SEA-CD40 (Seattle Genetics); and Chi Lob 7/4 (Univ. Southampton). In some embodiments, a CD40 agonist is recombinant CD40L.

Routes of Administration and Carriers



[0204] In various embodiments, antibodies may be administered in vivo by various routes, including, but not limited to, oral, intra-arterial, parenteral, intranasal, intramuscular, intracardiac, intraventricular, intratracheal, buccal, rectal, intraperitoneal, intradermal, topical, transdermal, and intrathecal, or otherwise by implantation or inhalation. The subject compositions may be formulated into preparations in solid, semi-solid, liquid, or gaseous forms; including, but not limited to, tablets, capsules, powders, granules, ointments, solutions, suppositories, enemas, injections, inhalants, and aerosols. A nucleic acid molecule encoding an antibody may be coated onto gold microparticles and delivered intradermally by a particle bombardment device, or "gene gun," as described in the literature (see, e.g., Tang et al., Nature 356:152-154 (1992)). The appropriate formulation and route of administration may be selected according to the intended application.

[0205] In various embodiments, compositions comprising antibodies are provided in formulations with a wide variety of pharmaceutically acceptable carriers (see, e.g., Gennaro, Remington: The Science and Practice of Pharmacy with Facts and Comparisons: Drugfacts Plus, 20th ed. (2003); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th ed., Lippencott Williams and Wilkins (2004); Kibbe et al., Handbook of Pharmaceutical Excipients, 3rd ed., Pharmaceutical Press (2000)). Various pharmaceutically acceptable carriers, which include vehicles, adjuvants, and diluents, are available. Moreover, various pharmaceutically acceptable auxiliary substances, such as Ph adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are also available. Non-limiting exemplary carriers include saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.

[0206] In various embodiments, compositions comprising antibodies may be formulated for injection, including subcutaneous administration, by dissolving, suspending, or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids, or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives. In various embodiments, the compositions may be formulated for inhalation, for example, using pressurized acceptable propellants such as dichlorodifluoromethane, propane, nitrogen, and the like. The compositions may also be formulated, in various embodiments, into sustained release microcapsules, such as with biodegradable or non-biodegradable polymers. A non-limiting exemplary biodegradable formulation includes poly lactic acid-glycolic acid polymer. A non-limiting exemplary non-biodegradable formulation includes a polyglycerin fatty acid ester. Certain methods of making such formulations are described, for example, in EP 1 125 584 A1.

[0207] Pharmaceutical packs and kits comprising one or more containers, each containing one or more doses of an antibody or combination of antibodies are also provided. In some embodiments, a unit dosage is provided wherein the unit dosage contains a predetermined amount of a composition comprising an antibody or combination of antibodies, with or without one or more additional agents. In some embodiments, such a unit dosage is supplied in single-use prefilled syringe for injection. In various embodiments, the composition contained in the unit dosage may comprise saline, sucrose, or the like; a buffer, such as phosphate, or the like; and/or be formulated within a stable and effective Ph range. Alternatively, in some embodiments, the composition may be provided as a lyophilized powder that may be reconstituted upon addition of an appropriate liquid, for example, sterile water. In some embodiments, the composition comprises one or more substances that inhibit protein aggregation, including, but not limited to, sucrose and arginine. In some embodiments, a composition of the invention comprises heparin and/or a proteoglycan.

[0208] Pharmaceutical compositions are administered in an amount effective for treatment or prophylaxis of the specific indication. The therapeutically effective amount is typically dependent on the weight of the subject being treated, his or her physical or health condition, the extensiveness of the condition to be treated, or the age of the subject being treated. In general, antibodies may be administered in an amount in the range of about 10 µg/kg body weight to about 100 mg/kg body weight per dose. In some embodiments, antibodies may be administered in an amount in the range of about 50 µg/kg body weight to about 5 mg/kg body weight per dose. In some embodiments, antibodies may be administered in an amount in the range of about 100 µg/kg body weight to about 10 mg/kg body weight per dose. In some embodiments, antibodies may be administered in an amount in the range of about 100 µg/kg body weight to about 20 mg/kg body weight per dose. In some embodiments, antibodies may be administered in an amount in the range of about 0.5 mg/kg body weight to about 20 mg/kg body weight per dose.

[0209] The antibody compositions may be administered as needed to subjects. Determination of the frequency of administration may be made by persons skilled in the art, such as an attending physician based on considerations of the condition being treated, age of the subject being treated, severity of the condition being treated, general state of health of the subject being treated and the like. In some embodiments, an effective dose of an antibody is administered to a subject one or more times. In various embodiments, an effective dose of an antibody is administered to the subject once a month, less than once a month, such as, for example, every two months or every three months. In other embodiments, an effective dose of an antibody is administered more than once a month, such as, for example, every three weeks, every two weeks or every week. In some embodiments, an effective dose of an antibody is administered once per 1, 2, 3, 4, or 5 weeks. In some embodiments, an effective dose of an antibody is administered twice or three times per week. An effective dose of an antibody is administered to the subject at least once. In some embodiments, the effective dose of an antibody may be administered multiple times, including for periods of at least a month, at least six months, or at least a year.

Additional Combination Therapy



[0210] The above therapeutic combinations may be administered alone or with other modes of treatment. They may be provided before, substantially contemporaneous with, or after other modes of treatment, for example, surgery, chemotherapy, radiation therapy, or the administration of a biologic, such as another therapeutic antibody. In some embodiments, the cancer has recurred or progressed following a therapy selected from surgery, chemotherapy, and radiation therapy, or a combination thereof.

[0211] For treatment of cancer, the combinations may be administered in conjunction with one or more additional anti-cancer agents, such as a chemotherapeutic agent, growth inhibitory agent, anti-cancer vaccine such as a gene therapy vaccine, anti-angiogenesis agent and/or anti-neoplastic composition. Nonlimiting examples of chemotherapeutic agent, growth inhibitory agent, anti-cancer vaccine, anti-angiogenesis agent and anti-neoplastic composition that can be used in combination with the antibodies of the present invention are provided herein under "Definitions."

[0212] In some embodiments, an anti-inflammatory drug may be administered with the combination, such as a steroid or a non-steroidal anti-inflammatory drug (NSAID).

EXAMPLES



[0213] The examples discussed below are intended to be purely exemplary of the invention and should not be considered to limit the invention in any way. The examples are not intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (for example, amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric.

Example 1: Humanized anti-CSFIR antibodies



[0214] Various humanized anti-CSFIR antibodies were developed previously. See, e.g., PCT Publication No. WO 2011/140249.

[0215] The sequences for each of the humanized heavy chain variable regions and humanized light chain variable regions, aligned with the sequences of the parental chimeric antibody variable regions and the sequences of the human acceptor variable framework regions are shown in Figures 1 (heavy chains) and 2 (light chains). The changes in humanized variable region sequences relative to the human acceptor variable framework region sequences are boxed. Each of the CDRs for each of the variable regions is shown in a boxed region, and labeled as "CDR" above the boxed sequences.

[0216] Table 8, below, shows the full sequences for the humanized heavy chains and humanized light chains of antibodies huAb1 to huAb16. The name and SEQ ID Nos of the humanized heavy chain and humanized light chain of each of those antibodies is shown in Table 3.
Table 3: Humanized heavy chains and light chains of huAb1 to huAb16
Humanized antibodyHumanized HCSEQ ID NOHumanized LCSEQ ID NO
huAb 1 h0301-H0 53 h0301-L0 60
huAb2 h0301-H1 54 h0301-L0 60
huAb3 h0301-H2 55 h0301-L0 60
huAb4 h0301-H0 53 h0301-L1 61
huAb5 h0301-H1 54 h0301-L1 61
huAb6 h0301-H2 55 h0301-L1 61
huAb7 h0302-H1 56 h0302-L0 62
huAb8 h0302-H1 56 h0302-L1 63
huAb9 h0302-H1 56 h0302-L2 64
huAb 10 h0302-H2 57 h0302-L0 62
huAb 11 h0302-H2 57 h0302-L1 63
huAb 12 h0302-H2 57 h0302-L2 64
huAb13 h0311-H1 58 h0311-L0 65
huAb 14 h0311-H1 58 h0311-L1 66
huAb 15 h0311-H2 59 h0311-L0 65
huAb 16 h0311-H2 59 h0311-L1 66


[0217] The 16 humanized antibodies were tested for binding to human, cynomolgus monkey, and mouse CSF1R ECD, as described previously. See, e.g., PCT Publication No. WO 2011/140249. The antibodies were found to bind to both human and cynomolgus monkey CSF1R ECD, but not to mouse CSF1R ECD. The humanized antibodies were also found to block binding of CSF1 and IL-34 to both human and cynomolgus CSF1R and to inhibit CSF1-induced and IL-34-induced phosphorylation of human CSF1R expressed in CHO cells. See, e.g., PCT Publication No. WO 2011/140249.

[0218] The ka, kd, and KD for binding to human CSF1R ECD were previously determined and are shown in Table 4. See, e.g., PCT Publication No. WO 2011/140249.
Table 4: Humanized antibody binding affinity for human CSF1R
huAbka(M-1s-1)Kd (s-1)KD (Nm)
huAb 0301-L0H0 3.22 x 106 1.11 x 10-03 0.35
huAb 0301-L0H1 3.56 x 106 1.22 x 10-03 0.34
huAb 0301-L0H2 2.32 x 106 6.60 x 10-04 0.28
huAb 0301-L1H0 3.29 x 106 1.15 x 10-03 0.35
huAb 0301-L1H1 2.87 x 106 9.21 x 10-04 0.32
huAb 0301-L1H2 2.95 x 106 7.42 x 10-04 0.25
huAb 0302-L0H1 3.54 x 106 3.69 x 10-03 1.04
huAb 0302-L1H1 3.47 x 106 4.04 x 10-03 1.17
huAb 0302-L2H1 1.60 x 106 9.14 x 10-04 0.57
huAb 0302-L0H2 3.40 x 106 1.79 x 10-03 0.53
huAb 0302-L1H2 2.71 x 106 1.53 x 10-03 0.56
huAb 0302-L2H2 1.84 x 106 8.40 x 10-04 0.46
huAb 0311-L0H1 1.22 x 106 5.40 x 10-04 0.44
huAb 0311-L1H1 1.32 x 106 6.64 x 10-04 0.50
huAb 0311-L0H2 1.34 x 106 4.73 x 10-04 0.35
huAb 0311-L1H2 1.51 x 106 6.09 x 10-04 0.40

Example 2: Enhancement of Anti-Tumor Activity with Combination Therapy



[0219] 6-8 week old female C57BL/6 mice are housed 5 animals per cage with access to food and water ad libitum. Mice are acclimated for at least 3 days after arrival in the vivarium. Mice are weighed and their flanks shaved prior to tumor cell line inoculation.

[0220] Murine colon adenocarcinoma cell ine MC38 is cultured in RPMI + 10 % FBS + 2 mM L-glutamine + antibiotic/antimycotic at 37°C with 5% CO2. Cells are suspended in a solution of 50%/v of DPBS and 50%/v of Matrigel at a concentration of 5 million cells/ml. 100 µl of cell solution (0.5 million cells) are implanted on the right flank of the each mouse using a 27G1/2 needle. Cells are kept from settling to the bottom of the tube by using an 18G needle and syringe and by slight vortex. Mice are anesthetized using isofluorane to reduce stress and to allow for more precise tumor cell implantation. Length (L) and Width (W) of each tumor is measured using an electronic caliper and the Volume (V) of the tumor will be calculated using V= (L x W2)/2. Once the mean tumor volume reaches approximately 105 mm3, mice are grouped and dosed as discussed below.

Groups and dosing



[0221] Mice were separated into the following groups and administered chimeric rat anti-mouse CSF1R antibody (mouse IgG1; referred to as "cmFPA008") and/or anti-CD40 antibody FGK45 (Bio X Cell; see Rolink et al., 1996, Immunity 5:319-330) according to the dosing schedules described below.
  • Control: Murine IgG, 30 mg/kg, i.p. 1x/wk, starting on day 0. Rat IgG 100 µg i.p. day 0.
  • huAb1: anti-CSFIR antibody, 30 mg/kg, i.p. 1x/wk, starting on day 0. Rat IgG 100 µg i.p. day 0.
  • Anti-CD40 (High): anti-CD40 antibody, 100 µg i.p. on day 0. Murine IgG, 30 mg/kg, i.p. 1x/wk, starting day 0.
  • Combo (High): anti-CSFIR antibody, 30 mg/kg, i.p. 1x/wk, starting on day 0, Anti-CD40, 100 µg i.p. day 0.
  • Anti-CD40 (Low): anti-CD40 antibody, 30 µg i.p. on day 0. Murine IgG, 30 mg/kg, i.p. 1x/wk, starting day 0.
  • Combo (Low): anti-CSFIR antibody, 30 mg/kg, i.p. 1x/wk, starting on day 0, anti-CD40 antibody, 30 µg i.p. day 0.


[0222] The dosing schedule and groupings are shown in Table 5:
Table 5: Study dosing and grouping
GroupTreatment #1: chimeric Ab1Dosing (mg/kg, schedule)Treatment #2: Anti-CD40Dosing (mg/kg, schedule)Mice (n)
1 Murine IgG 30, 1x week (qw) Rat IgG 100 µg on Day 0 10
2 anti-CSFIR antibody 10
3 Murine IgG anti-CD40 antibody 100 µg on Day 0 10
4 anti-CSFIR antibody 10
5 Murine IgG 30 µg on Day 0 10
6 anti-CSFIR antibody 10


[0223] Animals are euthanized prior to the end of the study if any of the following signs are observed:
  • Body weight loss of equal or greater than 15% of initial body weight.
  • Tumor ulceration is observed.
  • Mice appear moribund.
  • Individual tumor volume is equal or larger than 10% of initial body weight


[0224] Plasma is collected for pharmacokinetic (PK) analysis. On the final day of the study, whole blood is collected via intra-cardiac bleeds, and plasma is isolated for PK analysis (bioanalytical group). At least five (5) tumors from each group are collected for the following analyses. Single-cell isolates of the tumors are generated by collagenase treatment, and FACS is used to examine the infiltration of immune cells into the tumor(s). Tumor sections are also snap frozen in liquid nitrogen and stored at -80°C for protein and mRNA extraction. Tumor sections are embedded in Optimum Cutting Temperature compound (OCT) and stored in - 80°C. Tumor sections are also placed in 10% buffered formalin overnight, and then transferred to 70% ethanol the following day.

[0225] The results of this experiment are shown in Figures 3 and 4. As shown in Figure 3, the combination of anti-CSFIR antibody and anti-CD40 antibody (high) resulted in early tumor regression followed by tumor stasis. The combination of anti-CSF1R antibody and anti-CD40 antibody (low) also demonstrated greater efficacy than either treatment alone. Figure 4 shows individual tumor volumes at (A) day 11 and (B) day 13. Table 6 shows the tumor volume statistics at day 11 and day 13 using one-way ANOVA. The combination of anti-CSFIR antibody and anti-CD40 antibody (high) showed significantly better efficacy than either treatment alone.
Table 6: Tumor volume statistics
 Day 11Day 13
GroupMean TV±SD%TGIp-valueMean TV±SD%TGIp-value
IgG/IgG 729.3 ± 183.8 0.00 - 942.4 ± 278.3 0.00 -
anti-CSFIR antibody /IgG vs Combo(Low) 579.2 ± 87.87 20.58 0.0037 710.0 ± 80.05 24.66 0.0004
IgG/anti-CD40 antibody (low) vs Combo(Low) 452.5 ± 111.9 37.95 0.5888 ns 645.2 ± 162.0 31.53 0.0139
IgG/anti-CD40 antibody (High) vs Combo(High) 450.8 ± 105.6 38.18 < 0.0001 650.6 ± 151.5 30.96 < 0.0001
Combo (High) vs anti-CSFIR antibody /IgG 157.4 ± 55.00 78.41 < 0.0001 223.3 ± 98.32 76.31 < 0.0001


[0226] Figure 5 shows body weight for all animals in the study, measured at least twice per week. No significant different in weight was observed for any group relative to control.

TABLE OF SEQUENCES



[0227] Table 10 provides certain sequences discussed herein. All polypeptide and antibody sequences are shown without leader sequences, unless otherwise indicated.
Table 10: Sequences and Descriptions
SEQ ID NODescriptionSequence
1 hCSF1R (full-length, no leader sequence)

 
2 hCSF1R (full-length, + leader sequence)

 
5 hCSF1R ECD.506

 
   

 
6 hCSF1R ECD.506-Fc

 
7 cynoCSFIR ECD (with leader sequence)

 
8 cynoCSFIR ECD-Fc (with leader sequence)

 
3 Light chain leader sequence METDTLLLWV LLLWVPGSTG
4 Heavy chain leader sequence MAVLGLLLCL VTFPSCVLS
9 Fab 0301 heavy chain variable region

 
10 Fab 0301 light chain variable region

 
11 Fab 0302 heavy chain variable region

 
12 Fab 0302 light chain variable region

 
13 Fab 0311 heavy chain variable region

 
14 Fab 0311 light chain variable region

 
15 0301 heavy chain CDR1 GYTFTDNYMI
16 0301 heavy chain CDR2 DINPYNGGTT FNQKFKG
17 0301 heavy chain CDR3 ESPYFSNLYV MDY
18 0301 light chain CDR1 KASQSVDYDG DNYMN
19 0301 light chain CDR2 AASNLES
20 0301 light chain CDR3 HLSNEDLST
21 0302 heavy chain CDR1 GYTFSDFNIH
22 0302 heavy chain CDR2 YINPYTDVTV YNEKFKG
23 0302 heavy chain CDR3 YFDGTFDYAL DY
24 0302 light chain CDR1 RASESVDNYG LSFMN
25 0302 light chain CDR2 TASNLES
26 0302 light chain CDR3 QQSKELPWT
27 0311 heavy chain CDR1 GYIFTDYNMH
28 0311 heavy chain CDR2 EINPNNGVVV YNQKFKG
29 0311 heavy chain CDR3 ALYHSNFGWY FDS
30 0311 light chain CDR1 KASQSVDYDG DSHMN
31 0311 light chain CDR2 TASNLES
32 0311 light chain CDR3 QQGNEDPWT
33 cAb 0301 heavy chain

 
34 cAb 0301 light chain

 
35 cAb 0302 heavy chain

 
36 cAb 0302 light chain

 
37 cAb 0311 heavy chain

 
38 cAb 0311 light chain

 
39 h0301-H0 heavy chain variable region

 
40 h0301-H1 heavy chain variable region

 
41 h0301-H2 heavy chain variable region

 
42 H0302-H1 heavy chain variable region

 
43 H0302-H2 heavy chain variable region

 
44 H0311-H1 heavy chain variable region

 
45 H0311-H2 heavy chain variable region

 
46 h0301-L0 light chain variable region

 
47 h0301-L1 light chain variable region

 
48 H0302-L0 light chain variable region

 
49 H0302-L1 light chain variable region

 
50 H0302-L2 light chain variable region

 
51 H0311-L0 light chain variable region

 
52 H0311-L1 light chain variable region

 
53 h0301-H0 heavy chain

 
54 h0301-H1 heavy chain

 
55 h0301-H2 heavy chain

 
56 H0302-H1 heavy chain

 
57 H0302-H2 heavy chain

 
58 H0311-H1 heavy chain

 
59 H0311-H2 heavy chain

 
60 h0301-L0 light chain

 
   

 
61 h0301-L1 light chain

 
62 H0302-L0 light chain

 
63 H0302-L1 light chain

 
64 H0302-L2 light chain

 
65 H0311-L0 light chain

 
66 H0311-L1 light chain

 
67 Human CSF1

 
68 Human IL-34

 
69 Human acceptor A FR1 QVQLVQSGAE VKKPGSSVKV SCKAS
70 Human acceptor A FR2 WVRQAPGQGL EWMG
71 Human acceptor A FR3 RVTITADKST STAYMELSSL RSEDTAVYYC AR
72 Human acceptor A FR4 WGQGTLVTVS S
73 Human acceptor B FR1 QVQLVQSGAE VKKPGSSVKV SCKAS
74 Human acceptor B FR2 WVRQAPGQGL EWMG
75 Human acceptor B FR3 RVTITADKST STAYMELSSL RSEDTAVYYC AR
76 Human acceptor B FR4 WGQGTLVTVSS
77 Human acceptor C FR1 QVQLVQSGAE VKKPGSSVKV SCKAS
78 Human acceptor C FR2 WVRQAPGQGL EWMG
79 Human acceptor C FR3 RVTITADKST STAYMELSSL RSEDTAVYYC AR
80 Human acceptor C FR4 WGQGTLVTVS S
81 Human acceptor D FR1 EIVLTQSPAT LSLSPGERAT LSC
82 Human acceptor D FR2 WYQQKPGQAP RLLIY
83 Human acceptor D FR3 GIPARFSGSG SGTDFTLTIS SLEPEDFAVY YC
84 Human acceptor D FR4 FGGGTKVEIK
85 Human acceptor E FR1 EIVLTQSPAT LSLSPGERAT LSC
86 Human acceptor E FR2 WYQQKPGQAP RLLIY
87 Human acceptor E FR3 GIPARFSGSG SGTDFTLTIS SLEPEDFAVY YC
88 Human acceptor E FR4 FGQGTKVEIK
89 Human acceptor F FR1 EIVLTQSPAT LSLSPGERAT LSC
90 Human acceptor F FR2 WYQQKPGQAP RLLIY
91 Human acceptor F FR3 GIPARFSGSG SGTDFTLTIS SLEPEDFAVY YC
92 Human acceptor F FR4 FGQGTKVEIK
93 mCSF1R ECD-Fc

 
94 Human IgG4 S241P

 
95 Human Igκ

 
96 human CD40 precursor (with signal sequence) UniProtKB/S wiss-Prot: P25942.1, 04-MAR-2015

 
97 human CD40 (mature, without signal sequence)

 
98 Dacetuzumab heavy chain

 
99 Dcetuzumab light chain

 

SEQUENCE LISTING



[0228] 

<110> FIVE PRIME THERAPEUTICS, INC.
MASTELLER, Emma
BRENNAN, Thomas
BELLOVIN, David
BAKER, Kevin
WONG, Brian

<120> COMBINATION THERAPY FOR CANCER

<130> 01134-0044-00PCT

<150> US 62/146,766
<151> 2015-04-13

<150> US 62/190,945
<151> 2015-07-10

<160> 99

<170> PatentIn version 3.5

<210> 1
<211> 953
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(953)
<223> hCSF1R (full-length, no leader sequence)

<400> 1









<210> 2
<211> 972
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(972)
<223> hCSF1R (full-length, + leader sequence)

<400> 2









<210> 3
<211> 20
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Light chain leader sequence

<400> 3

<210> 4
<211> 19
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Heavy chain leader sequence

<400> 4

<210> 5
<211> 487
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: hCSF1R ECD.506

<400> 5





<210> 6
<211> 719
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: hCSF1R ECD.506-Fc

<400> 6







<210> 7
<211> 506
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cynoCSF1R ECD (with leader sequence)

<400> 7





<210> 8
<211> 740
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cynoCSF1R ECD-Fc (with leader sequence)

<400> 8







<210> 9
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Fab 0301 heavy chain variable region

<400> 9



<210> 10
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Fab 0301 light chain variable region

<400> 10

<210> 11
<211> 121
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Fab 0302 heavy chain variable region

<400> 11



<210> 12
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Fab 0302 light chain variable region

<400> 12



<210> 13
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Fab 0311 heavy chain variable region

<400> 13

<210> 14
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Fab 0311 light chain variable region

<400> 14



<210> 15
<211> 10
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0301 heavy chain CDR1

<400> 15

<210> 16
<211> 17
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0301 heavy chain CDR2

<400> 16

<210> 17
<211> 13
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0301 heavy chain CDR3

<400> 17



<210> 18
<211> 15
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0301 light chain CDR1

<400> 18

<210> 19
<211> 7
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0301 light chain CDR2

<400> 19

<210> 20
<211> 9
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0301 light chain CDR3

<400> 20

<210> 21
<211> 10
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0302 heavy chain CDR1

<400> 21

<210> 22
<211> 17
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0302 heavy chain CDR2

<400> 22

<210> 23
<211> 12
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0302 heavy chain CDR3

<400> 23

<210> 24
<211> 15
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0302 light chain CDR1

<400> 24

<210> 25
<211> 7
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0302 light chain CDR2

<400> 25

<210> 26
<211> 9
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0302 light chain CDR3

<400> 26

<210> 27
<211> 10
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0311 heavy chain CDR1

<400> 27

<210> 28
<211> 17
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0311 heavy chain CDR2

<400> 28

<210> 29
<211> 13
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0311 heavy chain CDR3

<400> 29

<210> 30
<211> 15
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0311 light chain CDR1

<400> 30

<210> 31
<211> 7
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0311 light chain CDR2

<400> 31

<210> 32
<211> 9
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: 0311 light chain CDR3

<400> 32

<210> 33
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cAb 0301 heavy chain

<400> 33





<210> 34
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cAb 0301 light chain

<400> 34



<210> 35
<211> 448
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cAb 0302 heavy chain

<400> 35





<210> 36
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cAb 0302 light chain

<400> 36



<210> 37
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cAb 0311 heavy chain

<400> 37





<210> 38
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: cAb 0311 light chain

<400> 38



<210> 39
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-HO heavy chain variable region

<400> 39



<210> 40
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-H1 heavy chain variable region

<400> 40

<210> 41
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-H2 heavy chain variable region

<400> 41

<210> 42
<211> 121
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-H1 heavy chain variable region

<400> 42



<210> 43
<211> 121
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-H2 heavy chain variable region

<400> 43

<210> 44
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-H1 heavy chain variable region

<400> 44

<210> 45
<211> 122
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-H2 heavy chain variable region

<400> 45



<210> 46
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-LO light chain variable region

<400> 46

<210> 47
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-L1 light chain variable region

<400> 47

<210> 48
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-L0 light chain variable region

<400> 48



<210> 49
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-L1 light chain variable region

<400> 49

<210> 50
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-L2 light chain variable region

<400> 50



<210> 51
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-LO light chain variable region

<400> 51

<210> 52
<211> 111
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-L1 light chain variable region

<400> 52

<210> 53
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-HO heavy chain

<400> 53





<210> 54
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-H1 heavy chain

<400> 54





<210> 55
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-H2 heavy chain

<400> 55





<210> 56
<211> 448
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-H1 heavy chain

<400> 56





<210> 57
<211> 448
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-H2 heavy chain

<400> 57





<210> 58
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-H1 heavy chain

<400> 58





<210> 59
<211> 449
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-H2 heavy chain

<400> 59





<210> 60
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-LO light chain

<400> 60



<210> 61
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: h0301-L1 light chain

<400> 61



<210> 62
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-L0 light chain

<400> 62



<210> 63
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-L1 light chain

<400> 63



<210> 64
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0302-L2 light chain

<400> 64



<210> 65
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-LO light chain

<400> 65



<210> 66
<211> 218
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: H0311-L1 light chain

<400> 66



<210> 67
<211> 158
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(158)
<223> Human CSF1

<400> 67



<210> 68
<211> 222
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(222)
<223> Human IL-34

<400> 68



<210> 69
<211> 25
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor A FR1

<400> 69

<210> 70
<211> 14
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor A FR2

<400> 70

<210> 71
<211> 32
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor A FR3

<400> 71

<210> 72
<211> 11
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor A FR4

<400> 72

<210> 73
<211> 25
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor B FR1

<400> 73

<210> 74
<211> 14
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor B FR2

<400> 74

<210> 75
<211> 32
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor B FR3

<400> 75

<210> 76
<211> 11
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor B FR4

<400> 76

<210> 77
<211> 25
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor C FR1

<400> 77

<210> 78
<211> 14
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor C FR2

<400> 78

<210> 79
<211> 32
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor C FR3

<400> 79

<210> 80
<211> 11
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor C FR4

<400> 80

<210> 81
<211> 23
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor D FR1

<400> 81

<210> 82
<211> 15
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor D FR2

<400> 82

<210> 83
<211> 32
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor D FR3

<400> 83

<210> 84
<211> 10
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor D FR4

<400> 84

<210> 85
<211> 23
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor E FR1

<400> 85

<210> 86
<211> 15
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor E FR2

<400> 86

<210> 87
<211> 32
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor E FR3

<400> 87

<210> 88
<211> 10
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor E FR4

<400> 88

<210> 89
<211> 23
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor F FR1

<400> 89

<210> 90
<211> 15
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor F FR2

<400> 90

<210> 91
<211> 32
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor F FR3

<400> 91

<210> 92
<211> 10
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human acceptor F FR4

<400> 92

<210> 93
<211> 719
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: mCSF1R ECD-Fc

<400> 93







<210> 94
<211> 327
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Human IgG4 S241P

<400> 94





<210> 95
<211> 107
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(107)
<223> Human Ig?

<400> 95

<210> 96
<211> 277
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(277)
<223> human CD40 precursor (with signal sequence)

<400> 96



<210> 97
<211> 257
<212> PRT
<213> Homo sapiens

<220>
<221> misc_feature
<222> (1)..(257)
<223> human CD40 (mature, without signal sequence)

<400> 97



<210> 98
<211> 444
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Dacetuzumab heavy chain

<400> 98





<210> 99
<211> 219
<212> PRT
<213> Artificial Sequence

<220>
<223> Synthetic: Dacetuzumab light chain

<400> 99






Claims

1. An anti-CSFIR antibody for use in a method of treating cancer in a subject, the method comprising administering to the subject the anti-CSFIR antibody and at least one immune stimulating agent,
wherein the anti-CSFIR antibody binds human CSF1R and is selected from:

i) an antibody comprising a heavy chain comprising the sequence of SEQ ID NO: 39 and a light chain comprising the sequence of SEQ ID NO: 46;

ii) an antibody comprising a heavy chain comprising a heavy chain (HC) CDR1 having the sequence of SEQ ID NO: 15, an HC CDR2 having the sequence of SEQ ID NO: 16, and an HC CDR3 having the sequence of SEQ ID NO: 17, and a light chain comprising a light chain (LC) CDR1 having the sequence of SEQ ID NO: 18, a LC CDR2 having the sequence of SEQ ID NO: 19, and a LC CDR3 having the sequence of SEQ ID NO: 20; and

iii) an antibody comprising a heavy chain comprising the sequence of SEQ ID NO: 53 and a light chain comprising the sequence of SEQ ID NO: 60;

and wherein the at least one immune stimulating agent comprises an agonist anti-CD40 antibody.
 
2. At least one immune stimulating agent for use in a method of treating cancer in a subject, the method comprising administering to the subject the at least one immune stimulating agent and an anti-CSFIR antibody, wherein the anti-CSFIR antibody binds human CSF1R and is selected from:

i) an antibody comprising a heavy chain comprising the sequence of SEQ ID NO: 39 and a light chain comprising the sequence of SEQ ID NO: 46;

ii) an antibody comprising a heavy chain comprising a heavy chain (HC) CDR1 having the sequence of SEQ ID NO: 15, an HC CDR2 having the sequence of SEQ ID NO: 16, and an HC CDR3 having the sequence of SEQ ID NO: 17, and a light chain comprising a light chain (LC) CDR1 having the sequence of SEQ ID NO: 18, a LC CDR2 having the sequence of SEQ ID NO: 19, and a LC CDR3 having the sequence of SEQ ID NO: 20; and

iii) an antibody comprising a heavy chain comprising the sequence of SEQ ID NO: 53 and a light chain comprising the sequence of SEQ ID NO: 60;

and wherein the at least one immune stimulating agent comprises an agonist anti-CD40 antibody.
 
3. A composition for use in a method of treating cancer, the composition comprising an anti-CSFIR antibody and at least one immune stimulating agent, wherein the anti-CSFIR antibody binds human CSF1R and is selected from:

i) an antibody comprising a heavy chain comprising the sequence of SEQ ID NO: 39 and a light chain comprising the sequence of SEQ ID NO: 46;

ii) an antibody comprising a heavy chain comprising a heavy chain (HC) CDR1 having the sequence of SEQ ID NO: 15, an HC CDR2 having the sequence of SEQ ID NO: 16, and an HC CDR3 having the sequence of SEQ ID NO: 17, and a light chain comprising a light chain (LC) CDR1 having the sequence of SEQ ID NO: 18, a LC CDR2 having the sequence of SEQ ID NO: 19, and a LC CDR3 having the sequence of SEQ ID NO: 20; and

iii) an antibody comprising a heavy chain comprising the sequence of SEQ ID NO: 53 and a light chain comprising the sequence of SEQ ID NO: 60;

and wherein the at least one immune stimulating agent comprises an agonist anti-CD40 antibody.
 
4. The anti-CSFIR antibody for use of claim 1, or the at least one immune stimulating agent for use of claim 2, or the composition for use of claim 3, wherein the at least one immune stimulating agent further comprises at least one additional immune stimulating agent chosen from agents falling within one or more of the following categories:

a. an agonist of an immune stimulatory molecule, including a co-stimulatory molecule, such as an immune-stimulatory molecule found on a T cell or NK cell;

b. an antagonist of an immune inhibitory molecule, including a co-inhibitory molecule, such as an immune-stimulatory molecule found on a T cell or NK cell;

c. an antagonist of LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGFβ, VEGF, KIR, LAG-3, adenosine A2A receptor, PI3Kdelta, or IDO;

d. an agonist of B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFNα, STING, or a Toll-like receptor agonist such as a TLR2/4 agonist;

e. an agent that binds to a member of the B7 family of membrane-bound proteins such as B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), and B7-H6;

f. an agent that binds to a member of the TNF receptor family or a co-stimulatory or co-inhibitory molecule binding to a member of the TNF receptor family such as CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fnl4, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LTβR, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNFβ, TNFR2, TNFα, 1β2, FAS, FASL, RELT, DR6, TROY, or NGFβ;

g. an agent that antagonizes or inhibits a cytokine that inhibits T cell activation such as IL-6, IL-10, TGFβ, VEGF;

h. an agonist of a cytokine that stimulates T cell activation such as IL-2, IL-7, IL-12, IL-15, IL-21, and IFNα; and

i. an antagonist of a chemokine, such as CXCR2, CXCR4, CCR2, or CCR4.


 
5. The anti-CSFIR antibody for use, at least one immune stimulating agent for use, or composition for use of any preceding claim, wherein the agonist anti-CD40 antibody comprises the CDRs of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
 
6. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the agonist anti-CD40 antibody comprises the heavy chain and light chain variable regions of an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
 
7. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the agonist anti-CD40 antibody is an antibody selected from CP-870,893; dacetuzumab; SEA-CD40; ADC-1013; RO7009789; and Chi Lob 7/4.
 
8. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the cancer is selected from non-small cell lung cancer, melanoma, squamous cell carcinoma of the head and neck, ovarian cancer, pancreatic cancer, renal cell carcinoma, hepatocellular carcinoma, bladder cancer, endometrial cancer, Hodgkin's lymphoma, lung cancer, glioma, gioblastoma multiforme, colon cancer, breast cancer, bone cancer, skin cancer, uterince cancer, gastric cancer, stomach cancer, lymphoma, lymphocytic leukemia, multiple myeloma, prostate cancer, mesothelioma, and kidney cancer.
 
9. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the cancer is recurrent or progressive after a therapy selected from surgery, chemotherapy, radiation therapy, or a combination thereof.
 
10. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the anti-CSFIR antibody blocks binding of CSF1 and/or IL-34 to CSF1R and/or inhibits ligand-induced CSF1R phosphorylation in vitro.
 
11. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the anti-CSFIR antibody inhibits ligand-induced CSF1R phosphorylation in vitro.
 
12. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the anti-CSFIR antibody is a humanized antibody.
 
13. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein the anti-CSFIR antibody is selected from a Fab, an Fv, an scFv, a Fab', and a (Fab')2.
 
14. The anti-CSFIR antibody for use, at least one immune stimulating agent for use or composition for use of any preceding claim, wherein in the method of treating cancer:

a) the anti-CSFIR antibody and the agonist anti-CD40 antibody are administered concurrently or sequentially; or

b) the anti-CSFIR antibody and the agonist anti-CD40 antibody are administered concurrently; or

c) one or more doses of the agonist anti-CD40 antibody are administered prior to administering the anti-CSFIR antibody; or

d) one or more doses of the anti-CSFIR antibody are administered prior to administering the agonist anti-CD40 antibody.


 
15. The composition for use according to any one of claims 3 to 14, wherein each of the anti-CSFIR antibody and the at least one immune stimulating agent are each present in separate compartments or containers.
 
16. The composition for use of any one of claims 3 to 14, wherein the anti-CSFIR antibody and at least one immune stimulating agent are mixed or formulated together.
 


Ansprüche

1. Anti-CSFIR-Antikörper zur Verwendung in einem Verfahren zum Behandeln von Krebs in einem Subjekt, wobei das Verfahren Verabreichen des Anti-CSFIR-Antikörpers und mindestens eines die Immunität stimulierenden Mittels an das Subjekt umfasst,
wobei der Anti-CSFIR-Antikörper humane CSF1R bindet und ausgewählt ist aus:

i) einem Antikörper, umfassend eine Schwerkette, die die Sequenz von SEQ ID NO: 39 umfasst, und eine Leichtkette, die die Sequenz von SEQ ID NO: 46 umfasst;

ii) einem Antikörper, umfassend eine Schwerkette, die eine Schwerketten-(HC) CDR1 umfasst, die die Sequenz von SEQ ID NO: 15 aufweist, eine HC CDR2, die die Sequenz von SEQ ID NO 16 aufweist und eine HC CDR3, die die Sequenz von SEQ ID NO: 17 aufweist, und eine Leichtkette, die eine Leichtketten-(LC) CDR1 umfasst, die die Sequenz von SEQ ID NO: 18 aufweist, eine LC CDR2, die die Sequenz von SEQ ID NO: 19 aufweist und eine LC CDR3, die die Sequenz von SEQ ID NO: 20 aufweist; und

iii) einem Antikörper, umfassend eine Schwerkette, die die Sequenz von SEQ ID NO: 53 umfasst, und eine Leichtkette, die die Sequenz von SEQ ID NO: 60 umfasst;

und wobei das mindestens eine die Immunität stimulierende Mittel einen Agonisten Anti-CD40-Antikörper umfasst.
 
2. Mindestens ein die Immunität stimulierendes Mittel zur Verwendung in einem Verfahren zum Behandeln von Krebs in einem Subjekt, wobei das Verfahren Verabreichen des mindestens einen die Immunität stimulierenden Mittels und eines Anti-CSF1R-Antikörpers an das Subjekt umfasst, wobei der Anti-CSFIR-Antikörper humane CSF1R bindet und ausgewählt ist aus:

i) einem Antikörper, umfassend eine Schwerkette, die die Sequenz von SEQ ID NO: 39 umfasst, und eine Leichtkette, die die Sequenz von SEQ ID NO: 46 umfasst;

ii) einem Antikörper, umfassend eine Schwerkette, die eine Schwerketten-(HC) CDR1 umfasst, die die Sequenz von SEQ ID NO: 15 aufweist, eine HC CDR2, die die Sequenz von SEQ ID NO 16 aufweist, und eine HC CDR3, die die Sequenz von SEQ ID NO: 17 aufweist, und eine Leichtkette, die eine Leichtketten-(LC) CDR1 umfasst, die die Sequenz von SEQ ID NO: 18 aufweist, eine LC CDR2, die die Sequenz von SEQ ID NO: 19 aufweist, und eine LC CDR3, die die Sequenz von SEQ ID NO: 20 aufweist; und

iii) einem Antikörper, umfassend eine Schwerkette, die die Sequenz von SEQ ID NO: 53 umfasst, und eine Leichtkette, die die Sequenz von SEQ ID NO: 60 umfasst;

und wobei das mindestens eine die Immunität stimulierende Mittel einen Agonisten Anti-CD40-Antikörper umfasst.
 
3. Zusammensetzung zur Verwendung in einem Verfahren zum Behandeln von Krebs, wobei die Zusammensetzung einen Anti-CSFIR-Antikörper und mindestens ein die Immunität stimulierendes Mittel umfasst, wobei der Anti-CSFIR-Antikörper humane CSF1R bindet und ausgewählt ist aus:

i) einem Antikörper, umfassend eine Schwerkette, die die Sequenz von SEQ ID NO: 39 umfasst, und eine Leichtkette, die die Sequenz von SEQ ID NO: 46 umfasst;

ii) einem Antikörper, umfassend eine Schwerkette, die eine Schwerketten-(HC) CDR1 umfasst, die die Sequenz von SEQ ID NO: 15 aufweist, eine HC CDR2, die die Sequenz von SEQ ID NO 16 aufweist, und eine HC CDR3, die die Sequenz von SEQ ID NO: 17 aufweist, und eine Leichtkette, umfassend eine Leichtketten-(LC) CDR1, die die Sequenz von SEQ ID NO: 18 aufweist, eine LC CDR2, die die Sequenz von SEQ ID NO: 19 aufweist, und eine LC CDR3, die die Sequenz von SEQ ID NO: 20 aufweist; und

iii) einem Antikörper, umfassend eine Schwerkette, die die Sequenz von SEQ ID NO: 53 umfasst, und eine Leichtkette, die die Sequenz von SEQ ID NO: 60 umfasst;

und wobei das mindestens eine die Immunität stimulierende Mittel einen Agonisten Anti-CD40-Antikörper umfasst.
 
4. Anti-CSFIR-Antikörper zur Verwendung nach Anspruch 1, oder das mindestens eine die Immunität stimulierende Mittel zur Verwendung nach Anspruch 2 oder die Zusammensetzung zur Verwendung nach Anspruch 3, wobei das mindestens eine die Immunität stimulierende Mittel ferner mindestens ein zusätzliches die Immunität stimulierendes Mittel umfasst, das ausgewählt ist aus Mitteln, die in eine oder mehrere der folgenden Kategorien fallen:

a. ein Agonist eines immunstimulatorischen Moleküls, einschließend ein co-stimulatorisches Molekül, wie beispielsweise ein immunstimulatorisches Molekül, das auf einer T-Zelle oder NK-Zelle angetroffen wird;

b. ein Antagonist eines immuninhibitorischen Moleküls, einschließlich eines co-inhibitorischen Moleküls, wie beispielsweise ein immunstimulatorisches Molekül, das auf einer T-Zelle oder NK-Zelle angetroffen wird;

c. ein Antagonist von LAG-3, Galectin 1, Galectin 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGFβ, VEGF, KIR, LAG-3, Adenosin A2A-Rezeptor, PI3Kdelta oder IDO;

d. ein Agonist von B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFNα, STING oder ein Toll-like-Rezeptor-Agonist, wie beispielsweise ein TLR2/4-Agonist;

e. ein Mittel, das an einem Vertreter der B7-Familie von Membrangebundenen Proteinen bindet, wie beispielsweise B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA), und B7-H6;

f. ein Mittel, das an einem Vertreter der TNF-Rezeptor-Familie bindet, oder ein co-stimulatorisches oder co-inhibitorisches Molekül, das an einem Vertreter der TNF-Rezeptor-Familie bindet, wie beispielsweise CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LTβR, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNFβ, TNFR2, TNFα, 1β2, FAS, FASL, RELT, DR6, TROY oder NGFβ;

g. ein Mittel, das einem Cytokin entgegenwirkt oder dieses hemmt, das T-Zell-Aktivierung hemmt, wie beispielsweise IL-6, IL-10, TGFβ, VEGF;

h. ein Agonist eines Cytokins, das T-Zell-Aktivierung stimuliert, wie beispielsweise IL-2, IL-7, IL-12, IL-15, IL-21, und IFNα; und

i. ein Antagonist eines Chemokins, wie beispielsweise CXCR2, CXCR4, CCR2 oder CCR4.


 
5. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Agonist Anti-CD40-Antikörper die CDR eines Antikörpers umfasst, der ausgewählt ist aus CP-870,893; Dacetuzumab; SEA-CD40; ADC-1013; RO7009789; und Chi Lob 7/4.
 
6. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Agonist Anti-CD40-Antikörper die Schwerketten- und Leichtketten-variablen Regionen eines Antikörpers umfasst, der ausgewählt ist aus CP-870,893; Dacetuzumab; SEA-CD40; ADC-1013; RO7009789; und Chi Lob 7/4.
 
7. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Agonist Anti-CD40-Antikörper ein Antikörper ist, der ausgewählt ist aus CP-870,893; Dacetuzumab; SEA-CD40; ADC-1013; RO7009789; und Chi Lob 7/4.
 
8. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Krebs ausgewählt ist aus: Nichtkleinzelligem Lungenkrebs, Melanom, Plattenepithelkarzinom des Kopfes und Halses, Eierstockkrebs, Bauchspeicheldrüsenkrebs, Nierenzellkarzinom, hepatozellulärem Karzinom, Blasenkrebs, Krebs des Endometriums, Hodgkin-Lymphom, Lungenkrebs, Gliom, Glioblastoma multiforme, Dickdarmkrebs, Brustkrebs, Knochenkrebs, Hautkrebs, Gebärmutterkrebs, gastrisches Karzinom, Magenkrebs, Lymphom, lymphatische Leukämie, Multiples Myelom, Prostatakrebs, Mesotheliom und Nierenkrebs.
 
9. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Krebs nach einer Therapie, die ausgewählt ist aus einer Operation, Chemotherapie, Strahlentherapie oder einer Kombination davon, rezidivierend oder fortschreitend ist.
 
10. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Anti-CSFIR-Antikörper Binden von CSF1 und/oder IL-34 an CSF1R blockiert und/oder durch Liganden eingeleitete Phosphorylierung in vitro von CSF1R hemmt.
 
11. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Anti-CSFIR-Antikörper durch Liganden eingeleitete Phosphorylierung in vitro des CSF1R hemmt.
 
12. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Anti-CSFIR-Antikörper ein humanisierter Antikörper ist.
 
13. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei der Anti-CSFIR-Antikörper ausgewählt ist aus einem Fab, einem Fv, einem scFv, einem Fab', und einem (Fab')2.
 
14. Anti-CSFIR-Antikörper zur Verwendung, mindestens ein die Immunität stimulierendes Mittel zur Verwendung oder Zusammensetzung zur Verwendung nach einem der vorhergehenden Ansprüche, wobei in dem Verfahren zum Behandeln von Krebs:

a) der Anti-CSFIR-Antikörper und der Agonist Anti-CD40-Antikörper gleichzeitig oder sequentiell verabreicht werden; oder

b) der Anti-CSF1R-Antikörper und der Agonist Anti-CD40-Antikörper gleichzeitig verabreicht werden; oder

c) eine oder mehrere Dosierungen des Agonist Anti-CD40-Antikörpers vor dem Verabreichen des Anti-CSFIR-Antikörpers verabreicht werden; oder

d) eine oder mehrere Dosierungen des Anti-CSFIR-Antikörpers vor dem Verabreichen des Agonisten Anti-CD40-Antikörper verabreicht werden.


 
15. Zusammensetzung zu Verwendung nach einem der Ansprüche 3 bis 14, wobei jeder der Anti-CSFIR-Antikörper und des mindestens einen die Immunität stimulierenden Mittels jeweils in separaten Kompartimenten oder Containern vorliegen.
 
16. Zusammensetzung zu Verwendung nach einem der Ansprüche 3 bis 14, wobei der Anti-CSFIR-Antikörper und mindestens ein die Immunität stimulierendes Mittel gemischt oder gemeinsam formuliert werden.
 


Revendications

1. Anticorps anti-CSF1R pour utilisation dans un procédé de traitement d'un cancer chez un sujet, le procédé comprenant l'administration au sujet de l'anticorps anti-CSF1R et d'au moins un agent stimulant le système immunitaire,
où l'anticorps anti-CSF1R se lie au CSF1R humain et est sélectionné parmi:

i) un anticorps comprenant une chaîne lourde comprenant la séquence de la SEQ ID NO: 39 et une chaîne légère comprenant la séquence de la SEQ ID NO: 46;

ii) un anticorps comprenant une chaîne lourde comprenant une CDR1 de chaîne lourde (HC) ayant la séquence de la SEQ ID NO: 15, une HC CDR2 ayant la séquence de la SEQ ID NO: 16, et une HC CDR3 ayant la séquence de la SEQ ID NO: 17, et une chaîne légère comprenant une CDR1 de chaîne légère (LC) ayant la séquence de la SEQ ID NO: 18, une LC CDR2 ayant la séquence de la SEQ ID NO: 19, et une LC CDR3 ayant la séquence de la SEQ ID NO: 20; et

iii) un anticorps comprenant une chaîne lourde comprenant la séquence de la SEQ ID NO: 53 et une chaîne légère comprenant la séquence de la SEQ ID NO: 60;

et où le au moins un agent stimulant le système immunitaire comprend un anticorps agoniste anti-CD40.
 
2. Au moins un agent stimulant le système immunitaire pour utilisation dans une procédé de traitement d'un cancer chez un sujet, le procédé comprenant l'administration au sujet de l'au moins un agent stimulant le système immunitaire et d'un anticorps anti-CSF1R, où l'anticorps anti-CSFIR se lie au CSF1R humain et est sélectionné parmi:

i) un anticorps comprenant une chaîne lourde comprenant la séquence de la SEQ ID NO: 39 et une chaîne légère comprenant la séquence de la SEQ ID NO: 46;

ii) un anticorps comprenant une chaîne lourde comprenant une CDR1 de chaîne lourde (HC) ayant la séquence de la SEQ ID NO: 15, une HC CDR2 ayant la séquence de la SEQ ID NO: 16, et une HC CDR3 ayant la séquence de la SEQ ID NO: 17, et une chaîne légère comprenant une CDR1 de chaîne légère (LC) ayant la séquence de la SEQ ID NO: 18, une LC CDR2 ayant la séquence de la SEQ ID NO: 19, et une LC CDR3 ayant la séquence de la SEQ ID NO: 20; et

iii) un anticorps comprenant une chaîne lourde comprenant la séquence de la SEQ ID NO: 53 et une chaîne légère comprenant la séquence de la SEQ ID NO: 60;

et où le au moins un agent stimulant le système immunitaire comprend un anticorps agoniste anti-CD40.
 
3. Composition pour utilisation dans un procédé de traitement d'un cancer, la composition comprenant un anticorps anti-CSF1R et au moins un agent stimulant le système immunitaire, où l'anticorps anti-CSF1R se lie au CSF1R humain et est sélectionné parmi:

i) un anticorps comprenant une chaîne lourde comprenant la séquence de la SEQ ID NO: 39 et une chaîne légère comprenant la séquence de la SEQ ID NO: 46;

ii) un anticorps comprenant une chaîne lourde comprenant une CDR1 de chaîne lourde (HC) ayant la séquence de la SEQ ID NO: 15, une HC CDR2 ayant la séquence de la SEQ ID NO: 16, et une HC CDR3 ayant la séquence de la SEQ ID NO: 17, et une chaîne légère comprenant une CDR1 de chaîne légère (LC) ayant la séquence de la SEQ ID NO: 18, une LC CDR2 ayant la séquence de la SEQ ID NO: 19, et une LC CDR3 ayant la séquence de la SEQ ID NO: 20; et

iii) un anticorps comprenant une chaîne lourde comprenant la séquence de la SEQ ID NO: 53 et une chaîne légère comprenant la séquence de la SEQ ID NO: 60;

et où le au moins un agent stimulant le système immunitaire comprend un anticorps agoniste anti-CD40.
 
4. Anticorps anti-CSF1R pour utilisation selon la revendication 1, ou le au moins un agent stimulant le système immunitaire pour utilisation selon la revendication 2, ou la composition pour utilisation selon la revendication 3, où le au moins un agent stimulant le système immunitaire comprend en outre au moins un agent stimulant le système immunitaire supplémentaire sélectionné parmi des agents tombant dans une ou plusieurs des catégories suivantes:

a. un agoniste d'une molécule stimulant le système immunitaire, y compris une molécule co-stimulante, telle qu'une molécule stimulant le système immunitaire trouvée sur une cellule T ou une cellule NK;

b. un antagoniste d'une molécule inhibant le système immunitaire, y compris une molécule co-inhibitrice, telle qu'une molécule stimulant_le système immunitaire trouvée sur une cellule T ou une cellule NK;

c. un antagoniste de LAG-3, galectine 1, galectine 9, CEACAM-1, BTLA, CD25, CD69, TIGIT, CD113, GPR56, VISTA, B7-H3, B7-H4, 2B4, CD48, GARP, PD1H, LAIR1, TIM1, TIM3, TIM4, ILT4, IL-6, IL-10, TGFβ, VEGF, KIR, LAG-3, récepteur A2A de l'adénosine, PI3Kdelta ou IDO;

d. un agoniste de B7-1, B7-2, CD28, 4-1BB (CD137), 4-1BBL, ICOS, ICOS-L, OX40, OX40L, GITR, GITRL, CD27, CD40, CD40L, DR3, CD28H, IL-2, IL-7, IL-12, IL-15, IL-21, IFNα, STING, ou un agoniste du récepteur Toll-like tel qu'un agoniste de TLR2/4;

e. un agent qui se lie à un membre de la famille B7 de protéines liées à la membrane telles que B7-1, B7-2, B7-H2 (ICOS-L), B7-H3, B7-H4, B7-H5 (VISTA) et B7-H6;

f. un agent qui se lie à un membre de la famille des récepteurs du TNF ou une molécule co-stimulante ou co-inhibitrice se liant à un membre de la famille des récepteurs du TNF tel que CD40, CD40L, OX40, OX40L, GITR, GITRL, CD70, CD27L, CD30, CD30L, 4-1BBL, CD137 (4-1BB), TRAIL/Apo2-L, TRAILR1/DR4, TRAILR2/DR5, TRAILR3, TRAILR4, OPG, RANK, RANKL, TWEAKR/Fn14, TWEAK, BAFFR, EDAR, XEDAR, EDA1, EDA2, TACI, APRIL, BCMA, LTβR, LIGHT, DeR3, HVEM, VEGL/TL1A, TRAMP/DR3, TNFR1, TNFβ, TNFR2, TNFα, 1β2, FAS, FASL, RELT, DR6, TROY ou NGFβ;

g. un agent qui antagonise ou inhibe une cytokine qui inhibe l'activation des cellules T telle qu'IL-6, IL-10, TGFβ, VEGF;

h. un agoniste d'une cytokine qui stimule l'activation des cellules T telle qu'IL-2, IL-7, IL-12, IL-15, IL-21 et IFNα; et

i. un antagoniste d'une chimiokine, telle que CXCR2, CXCR4, CCR2 ou CCR4.


 
5. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps agoniste anti-CD40 comprend les CDR d'un anticorps sélectionné parmi CP-870,893; dacétuzumab; SEA-CD40; ADC-1013; RO7009789 et Chi Lob 7/4.
 
6. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps agoniste anti-CD40 comprend les régions variables de la chaîne lourde et de la chaîne légère d'un anticorps sélectionné parmi CP-870,893; dacétuzumab; SEA-CD40; ADC-1013; RO7009789 et Chi Lob 7/4.
 
7. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps agoniste anti-CD40 est un anticorps sélectionné parmi CP-870,893; dacétuzumab; SEA-CD40; ADC-1013; RO7009789 et Chi Lob 7/4.
 
8. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où le cancer est sélectionné parmi : cancer du poumon non à petites cellules, mélanome, carcinome à cellules squameuses de la tête et du cou, cancer de l'ovaire, cancer du pancréas, carcinome à cellules rénales, carcinome hépatocellulaire, cancer de la vessie, cancer de l'endomètre, lymphome hodgkinien, cancer du poumon, gliome, glioblastome multiforme, cancer du côlon, cancer du sein, cancer des os, cancer de la peau, cancer de l'utérus, cancer gastrique, cancer de l'estomac, lymphome, leucémie lymphocytaire, myélome multiple, cancer de la prostate, mésothéliome et cancer du rein.
 
9. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où le cancer est récurrent ou progressif suite à une thérapie sélectionnée parmi chirurgie, chimiothérapie, radiothérapie ou une combinaison de celles-ci.
 
10. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps anti-CSF1R bloque la liaison de CSF-1 et/ou d'IL-34 à CSF1R et/ou inhibe la phosphorylation de CSF1R induite par ligand in vitro.
 
11. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps anti-CSF1R inhibe la phosphorylation de CSF1R induite par ligand in vitro.
 
12. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps anti-CSF1R est un anticorps humanisé.
 
13. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où l'anticorps anti-CSF1R est sélectionné parmi un Fab, un Fv, un scFv, un Fab' et un (Fab')2.
 
14. Anticorps anti-CSF1R pour utilisation, au moins un agent stimulant le système immunitaire pour utilisation, ou une composition pour utilisation selon l'une quelconque des revendications précédentes, où dans le procédé de traitement d'un cancer:

a) l'anticorps anti-CSF1R et l'anticorps agoniste anti-CD40 sont administrés simultanément ou séquentiellement; ou

b) l'anticorps anti-CSF1R et l'anticorps agoniste anti-CD40 sont administrés simultanément; ou bien

c) une ou plusieurs doses de l'anticorps agoniste anti-CD40 sont administrées avant d'administrer l'anticorps anti-CSF1R; ou

d) une ou plusieurs doses de l'anticorps anti-CSF1R sont administrées avant d'administrer l'anticorps agoniste anti-CD40.


 
15. Composition pour utilisation selon l'une quelconque des revendications 3 à 14, dans laquelle chacun de l'anticorps anti-CSF1R et du au moins un agent stimulant le système immunitaire sont présents chacun dans des compartiments ou récipients séparés.
 
16. Composition pour utilisation selon l'une quelconque des revendications 3-14, dans laquelle l'anticorps anti-CSF1R et au moins un agent stimulant le système immunitaire sont mélangés ou formulés ensemble.
 




Drawing
































Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description