48 results on '"Weiskopf K"'
Search Results
2. SIRPalpha antibody complex
- Author
-
Ring, N.G., primary, Herndler-Brandstetter, D., additional, Weiskopf, K., additional, Shan, L., additional, Volkmer, J.P., additional, George, B.M., additional, Lietzenmayer, M., additional, McKenna, K.M., additional, Naik, T.J., additional, McCarty, A., additional, Zheng, Y., additional, Ring, A.M., additional, Flavell, R.A., additional, and Weissman, I.L., additional
- Published
- 2017
- Full Text
- View/download PDF
3. Asterless is a scaffold for the onset of centriole assembly
- Author
-
Dzhindzhev, Ns, Yu, Qd, Weiskopf, K., Tzolovsky, G., Cunha Ferreira, I., Riparbelli, Maria Giovanna, Rodrigues Martins, A., Bettencourt Dias, M., Giuliano CALLAINI, and Glover, Dm
- Published
- 2010
4. Macrophage Transplantation Improves Cutaneous Wound Healing in a Humanized Mouse Model
- Author
-
Walmsley, G.G., primary, Hu, M.S., additional, Weiskopf, K., additional, Rennert, R.C., additional, McArdle, A., additional, Chung, M.T., additional, Schnorr, P.J., additional, Rajadas, J., additional, Gurtner, G.C., additional, Weissman, I.L., additional, Lorenz, H.P., additional, and Longaker, M.T., additional
- Published
- 2014
- Full Text
- View/download PDF
5. CD137 stimulation of natural killer cells to enhance the antilymphoma activity of rituximab.
- Author
-
Kohrt, H. E., primary, Houot, R., additional, Goldstein, M., additional, Weiskopf, K., additional, Chen, L., additional, Tedder, T., additional, and Levy, R., additional
- Published
- 2010
- Full Text
- View/download PDF
6. CD47 predominates over CD24 as a macrophage immune checkpoint in cancer.
- Author
-
Allen J, Meglan A, Vaccaro K, Velarde J, Chen V, Ribeiro J, Blandin J, Gupta S, Mishra R, Ho R, Love J, Reinhardt F, Bell GW, Chen J, Weinberg R, Yang D, Weissman J, and Weiskopf K
- Abstract
Macrophages hold tremendous promise as effectors of cancer immunotherapy, but the best strategies to provoke these cells to attack tumors remain unknown. Here, we evaluated the therapeutic potential of targeting two distinct macrophage immune checkpoints: CD47 and CD24. We found that antibodies targeting these antigens could elicit maximal levels of phagocytosis when combined together in vitro. However, to our surprise, via unbiased genome-wide CRISPR screens, we found that CD24 primarily acts as a target of opsonization rather than an immune checkpoint. In a series of in vitro and in vivo genetic validation studies, we found that CD24 was neither necessary nor sufficient to protect cancer cells from macrophage phagocytosis in most mouse and human tumor models. Instead, anti-CD24 antibodies exhibit robust Fc-dependent activity, and as a consequence, they cause significant on-target hematologic toxicity in mice. To overcome these challenges and leverage our findings for therapeutic purposes, we engineered a collection of 77 novel bispecific antibodies that bind to a tumor antigen with one arm and engage macrophages with the second arm. We discovered multiple novel bispecifics that maximally activate macrophage-mediated cytotoxicity and reduce binding to healthy blood cells, including bispecifics targeting macrophage immune checkpoint molecules in combination with EGFR, TROP2, and CD71. Overall, our findings indicate that CD47 predominates over CD24 as a macrophage immune checkpoint in cancer, and that the novel bispecifics we created may be optimal immunotherapies to direct myeloid cells to eradicate solid tumors.
- Published
- 2024
- Full Text
- View/download PDF
7. Unbiased discovery of antibody therapies that stimulate macrophage-mediated destruction of B-cell lymphoma.
- Author
-
Ribeiro J, Pagès-Geli C, Meglan A, Velarde J, Blandin J, Vaccaro K, Wienclaw T, Fernández-Guzmán P, Hahn CK, Crespo M, and Weiskopf K
- Abstract
Macrophages are critical effectors of antibody therapies for lymphoma, but the best targets for this purpose remain unknown. Here, we sought to define a comprehensive repertoire of cell surface antigens that can be targeted to stimulate macrophage-mediated destruction of B-cell lymphoma. We developed a high-throughput assay to screen hundreds of antibodies for their ability to provoke macrophages to attack B-cell lymphoma cells. Across both mouse and human systems, we identified multiple unappreciated targets of opsonization as well as putative immune checkpoints. We used this information to engineer a compendium of 156 bispecific antibodies, and we identified dozens of bispecifics that dramatically stimulate macrophage-mediated cytotoxicity of lymphoma cells. Among these, a bispecific comprising a SIRPα decoy domain and a CD38-targeting arm (WTa2d1×CD38) exhibited maximal efficacy while minimizing the risk of hematologic toxicity. This bispecific stimulated robust anti-tumor responses in multiple xenograft models of aggressive B-cell lymphoma. Our approach can be directly applied to other cancers to rapidly discover bispecific antibodies that leverage anti-tumor responses by macrophages or other innate immune cells., Competing Interests: Competing interests JR, CPG, KV, AM, and KW have filed US patent applications related to this work. KV is a former employee and equity owner of DEM Biopharma. MC has received research funding from Janssen, Genentech and AstraZeneca. KW reports patents/royalties (Stanford University, Whitehead Institute, Forty Seven, Gilead Sciences, ALX Oncology, DEM Biopharma); co-founder, scientific advisory board member, and equity holder (ALX Oncology, DEM Biopharma, Solu Therapeutics), stock ownership (Ginkgo Bioworks). The other authors declare no competing interests.
- Published
- 2024
- Full Text
- View/download PDF
8. Targeted therapies prime oncogene-driven lung cancers for macrophage-mediated destruction.
- Author
-
Vaccaro K, Allen J, Whitfield TW, Maoz A, Reeves S, Velarde J, Yang D, Meglan A, Ribeiro J, Blandin J, Phan N, Bell GW, Hata AN, and Weiskopf K
- Subjects
- Humans, Mice, Animals, Cell Line, Tumor, Mutation, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Molecular Targeted Therapy, ErbB Receptors genetics, ErbB Receptors metabolism, ErbB Receptors antagonists & inhibitors, ErbB Receptors immunology, MAP Kinase Signaling System drug effects, MAP Kinase Signaling System immunology, MAP Kinase Signaling System genetics, Phagocytosis, Female, Lung Neoplasms genetics, Lung Neoplasms drug therapy, Lung Neoplasms pathology, Lung Neoplasms immunology, Lung Neoplasms metabolism, CD47 Antigen genetics, CD47 Antigen metabolism, CD47 Antigen immunology, CD47 Antigen antagonists & inhibitors, Macrophages metabolism, Macrophages immunology, Macrophages pathology
- Abstract
Macrophage immune checkpoint inhibitors, such as anti-CD47 antibodies, show promise in clinical trials for solid and hematologic malignancies. However, the best strategies to use these therapies remain unknown, and ongoing studies suggest they may be most effective when used in combination with other anticancer agents. Here, we developed an unbiased, high-throughput screening platform to identify drugs that render lung cancer cells more vulnerable to macrophage attack, and we found that therapeutic synergy exists between genotype-directed therapies and anti-CD47 antibodies. In validation studies, we found that the combination of genotype-directed therapies and CD47 blockade elicited robust phagocytosis and eliminated persister cells in vitro and maximized antitumor responses in vivo. Importantly, these findings broadly applied to lung cancers with various RTK/MAPK pathway alterations - including EGFR mutations, ALK fusions, or KRASG12C mutations. We observed downregulation of β2-microglobulin and CD73 as molecular mechanisms contributing to enhanced sensitivity to macrophage attack. Our findings demonstrate that dual inhibition of the RTK/MAPK pathway and the CD47/SIRPa axis is a promising immunotherapeutic strategy. Our study provides strong rationale for testing this therapeutic combination in patients with lung cancers bearing driver mutations.
- Published
- 2024
- Full Text
- View/download PDF
9. MMD collaborates with ACSL4 and MBOAT7 to promote polyunsaturated phosphatidylinositol remodeling and susceptibility to ferroptosis.
- Author
-
Phadnis VV, Snider J, Varadharajan V, Ramachandiran I, Deik AA, Lai ZW, Kunchok T, Eaton EN, Sebastiany C, Lyakisheva A, Vaccaro KD, Allen J, Yao Z, Wong V, Geng B, Weiskopf K, Clish CB, Brown JM, Stagljar I, Weinberg RA, and Henry WS
- Subjects
- Cell Line, Fatty Acids, Unsaturated, Phospholipids metabolism, Humans, Long-Chain-Fatty-Acid-CoA Ligase, Ferroptosis, Phosphatidylinositols metabolism
- Abstract
Ferroptosis is a form of regulated cell death with roles in degenerative diseases and cancer. Excessive iron-catalyzed peroxidation of membrane phospholipids, especially those containing the polyunsaturated fatty acid arachidonic acid (AA), is central in driving ferroptosis. Here, we reveal that an understudied Golgi-resident scaffold protein, MMD, promotes susceptibility to ferroptosis in ovarian and renal carcinoma cells in an ACSL4- and MBOAT7-dependent manner. Mechanistically, MMD physically interacts with both ACSL4 and MBOAT7, two enzymes that catalyze sequential steps to incorporate AA in phosphatidylinositol (PI) lipids. Thus, MMD increases the flux of AA into PI, resulting in heightened cellular levels of AA-PI and other AA-containing phospholipid species. This molecular mechanism points to a pro-ferroptotic role for MBOAT7 and AA-PI, with potential therapeutic implications, and reveals that MMD is an important regulator of cellular lipid metabolism., Competing Interests: Declaration of interests K.W. declares relationships pertaining to macrophage-directed therapies, including patents and royalties (Stanford University, Whitehead Institute, Gilead Sciences), is a co-founder, SAB member, and equity holder (ALX Oncology, DEM Biopharma), and is a scientific advisor (Carisma Therapeutics)., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Phagocytic cooperativity by tumour macrophages.
- Author
-
Maoz A and Weiskopf K
- Subjects
- Humans, Macrophages, Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
11. Targeted therapies prime oncogene-driven lung cancers for macrophage-mediated destruction.
- Author
-
Vaccaro K, Allen J, Whitfield TW, Maoz A, Reeves S, Velarde J, Yang D, Phan N, Bell GW, Hata AN, and Weiskopf K
- Abstract
Macrophage immune checkpoint inhibitors, such as anti-CD47 antibodies, show promise in clinical trials for solid and hematologic malignancies. However, the best strategies to use these therapies remain unknown and ongoing studies suggest they may be most effective when used in combination with other anticancer agents. Here, we developed a novel screening platform to identify drugs that render lung cancer cells more vulnerable to macrophage attack, and we identified therapeutic synergy exists between genotype-directed therapies and anti-CD47 antibodies. In validation studies, we found the combination of genotype-directed therapies and CD47 blockade elicited robust phagocytosis and eliminated persister cells in vitro and maximized anti-tumor responses in vivo. Importantly, these findings broadly applied to lung cancers with various RTK/MAPK pathway alterations-including EGFR mutations, ALK fusions, or KRAS
G12C mutations. We observed downregulation of β2-microglobulin and CD73 as molecular mechanisms contributing to enhanced sensitivity to macrophage attack. Our findings demonstrate that dual inhibition of the RTK/MAPK pathway and the CD47/SIRPa axis is a promising immunotherapeutic strategy. Our study provides strong rationale for testing this therapeutic combination in patients with lung cancers bearing driver mutations., Competing Interests: Conflict-of-interest statement: K.V. is currently an employee and equity owner of DEM Biopharma. D.Y. is a co-founder, SAB member, and equity holder of DEM Biopharma. N.P. is a current employee of Bristol-Myers Squibb. A.N.H. has received research support from Amgen, Blueprint Medicines, BridgeBio, Bristol-Myers Squibb, C4 Therapeutics, Eli Lilly, Nuvalent, Pfizer, Roche/Genentech, Scorpion Therapeutics; has served as a compensated consultant for Nuvalent, Tolremo Therapeutics, Engine Biosciences and TigaTx. K.W. declares relevant relationships pertaining to macrophage-directed therapies including patents and royalties (Stanford University, Whitehead Institute, Gilead Sciences); co-founder, SAB member, and equity holder (ALX Oncology, DEM Biopharma); scientific advisor (Carisma Therapeutics). K.W. reports stock ownership of Ginkgo Bioworks. K.W., A.H., K.D.V, J.L.A, D.Y., and A.M. have filed U.S. patent applications related to this work. The other authors have declared that no conflict of interest exists.- Published
- 2023
- Full Text
- View/download PDF
12. Evolutionarily conserved resistance to phagocytosis observed in melanoma cells is insensitive to upregulation of pro-phagocytic signals and to CD47 blockade.
- Author
-
Anderson KL, Snyder KM, Ito D, Lins DC, Mills LJ, Weiskopf K, Ring NG, Ring AM, Shimizu Y, Mescher MF, Weissman IL, and Modiano JF
- Subjects
- Animals, Cell Line, Tumor, Humans, Mice, Signal Transduction, Transfection, Up-Regulation, CD47 Antigen metabolism, Melanoma genetics, Phagocytosis physiology
- Abstract
Therapeutic activation of macrophage phagocytosis has the ability to restrain tumour growth through phagocytic clearance of tumour cells and activation of the adaptive immune response. Our objective for this study was to evaluate the effects of modulating pro- and anti-phagocytic pathways in malignant melanoma. In order to identify evolutionarily conserved mechanisms of resistance that may be important for melanoma cell survival, we utilized a multi-species approach and examined the phagocytosis of human, mouse, and dog melanoma cells. We observed that melanoma cells from all three species displayed unexpected resistance to phagocytosis that could not be fully mitigated by blockade of the 'don't eat me' signal CD47 or by chemotherapeutic enhancement of known 'eat me' signals. Additionally, CD47 blockade failed to promote anti-melanoma immune responses or tumour regression in vivo. This melanoma resistance to phagocytosis was not mediated by soluble factors, and it was unaffected by siRNA-mediated knockdown of 47 prospective 'don't eat me' signals or by CRISPR-Cas-mediated CD47 knockout. Unexpectedly, CD47 knockout also did not enhance phagocytosis of lymphoma cells, but it eliminated the pro-phagocytic effect of CD47 blockade, suggesting that the pro-phagocytic effects of CD47 blockade are due in part to Fc receptor engagement. From this study, we conclude that melanoma cells possess an evolutionarily conserved resistance to macrophage phagocytosis. Further investigation will be needed to overcome the mechanisms that mediate melanoma cell resistance to innate immunity.
- Published
- 2020
- Full Text
- View/download PDF
13. Identification of Racial Inequities in Access to Specialized Inpatient Heart Failure Care at an Academic Medical Center.
- Author
-
Eberly LA, Richterman A, Beckett AG, Wispelwey B, Marsh RH, Cleveland Manchanda EC, Chang CY, Glynn RJ, Brooks KC, Boxer R, Kakoza R, Goldsmith J, Loscalzo J, Morse M, Lewis EF, Abel S, Adams A, Anaya J, Andrews EH, Atkinson B, Avutu V, Bachorik A, Badri O, Bailey M, Baird K, Bakshi S, Balaban D, Barshop K, Baumrin E, Bayomy O, Beamesderfer J, Becker N, Berg DD, Berman AN, Blum SM, Boardman AP, Boden K, Bonacci RA, Brown S, Campbell K, Case S, Cetrone E, Charrow A, Chiang D, Clark D, Cohen AJ, Cooper A, Cordova T, Cuneo CN, de Feria AA, Deffenbacher K, DeFilippis EM, DeGregorio G, Deutsch AJ, Diephuis B, Divakaran S, Dorschner P, Downing N, Drescher C, D'Silva KM, Dunbar P, Duong D, Earp S, Eckhardt C, Elman SA, England R, Everett K, Fedotova N, Feingold-Link T, Ferreira M, Fisher H, Foo P, Foote M, Franco I, Gilliland T, Greb J, Greco K, Grewal S, Grin B, Growdon ME, Guercio B, Hahn CK, Hasselfeld B, Haydu EJ, Hermes Z, Hildick-Smith G, Holcomb Z, Holroyd K, Horton L, Huang G, Jablonski S, Jacobs D, Jain N, Japa S, Joseph R, Kalashnikova M, Kalwani N, Kang D, Karan A, Katz JT, Kellner D, Kidia K, Kim JH, Knowles SM, Kolbe L, Kore I, Koullias Y, Kuye I, Lang J, Lawlor M, Lechner MG, Lee K, Lee S, Lee Z, Limaye N, Lin-Beckford S, Lipsyc M, Little J, Loewenthal J, Logaraj R, Lopez DM, Loriaux D, Lu Y, Ma K, Marukian N, Matias W, Mayers JR, McConnell I, McLaughlin M, Meade C, Meador C, Mehta A, Messenger E, Michaelidis C, Mirsky J, Mitten E, Mueller A, Mullur J, Munir A, Murphy E, Nagami E, Natarajan A, Nsahlai M, Nze C, Okwara N, Olds P, Paez R, Pardo M, Patel S, Petersen A, Phelan L, Pimenta E, Pipilas D, Plovanich M, Pong D, Powers BW, Rao A, Ramirez Batlle H, Ramsis M, Reichardt A, Reiger S, Rengarajan M, Rico S, Rome BN, Rosales R, Rotenstein L, Roy A, Royston S, Rozansky H, Rudder M, Ryan CE, Salgado S, Sanchez P, Schulte J, Sekar A, Semenkovich N, Shannon E, Shaw N, Shorten AB, Shrauner W, Sinnenberg L, Smithy JW, Snyder G, Sreekrishnan A, Stabenau H, Stavrou E, Stergachis A, Stern R, Stone A, Tabrizi S, Tanyos S, Thomas C, Thun H, Torres-Lockhart K, Tran A, Treasure C, Tsai FD, Tsaur S, Tschirhart E, Tuwatananurak J, Venkateswaran RV, Vishnevetsky A, Wahl L, Wall A, Wallace F, Walsh E, Wang P, Ward HB, Warner LN, Weeks LD, Weiskopf K, Wengrod J, Williams JN, Winkler M, Wong JL, Worster D, Wright A, Wunsch C, Wynter JS, Yarbrough C, Yau WY, Yazdi D, Yeh J, Yialamas MA, Yozamp N, Zambrotta M, and Zon R
- Subjects
- Aged, Aged, 80 and over, Boston epidemiology, Female, Health Status Disparities, Heart Failure diagnosis, Heart Failure ethnology, Heart Failure mortality, Humans, Inpatients, Male, Middle Aged, Patient Readmission, Retrospective Studies, Risk Assessment, Risk Factors, Time Factors, Treatment Outcome, Academic Medical Centers, Black or African American, Cardiology Service, Hospital, Health Services Accessibility, Healthcare Disparities ethnology, Heart Failure therapy, Hispanic or Latino, Patient Admission, White People
- Abstract
Background: Racial inequities for patients with heart failure (HF) have been widely documented. HF patients who receive cardiology care during a hospital admission have better outcomes. It is unknown whether there are differences in admission to a cardiology or general medicine service by race. This study examined the relationship between race and admission service, and its effect on 30-day readmission and mortality Methods: We performed a retrospective cohort study from September 2008 to November 2017 at a single large urban academic referral center of all patients self-referred to the emergency department and admitted to either the cardiology or general medicine service with a principal diagnosis of HF, who self-identified as white, black, or Latinx. We used multivariable generalized estimating equation models to assess the relationship between race and admission to the cardiology service. We used Cox regression to assess the association between race, admission service, and 30-day readmission and mortality., Results: Among 1967 unique patients (66.7% white, 23.6% black, and 9.7% Latinx), black and Latinx patients had lower rates of admission to the cardiology service than white patients (adjusted rate ratio, 0.91; 95% CI, 0.84-0.98, for black; adjusted rate ratio, 0.83; 95% CI, 0.72-0.97 for Latinx). Female sex and age >75 years were also independently associated with lower rates of admission to the cardiology service. Admission to the cardiology service was independently associated with decreased readmission within 30 days, independent of race., Conclusions: Black and Latinx patients were less likely to be admitted to cardiology for HF care. This inequity may, in part, drive racial inequities in HF outcomes.
- Published
- 2019
- Full Text
- View/download PDF
14. Targeting CD137 enhances the efficacy of cetuximab.
- Author
-
Kohrt HE, Colevas AD, Houot R, Weiskopf K, Goldstein MJ, Lund P, Mueller A, Sagiv-Barfi I, Marabelle A, Lira R, Troutner E, Richards L, Rajapaska A, Hebb J, Chester C, Waller E, Ostashko A, Weng WK, Chen L, Czerwinski D, Fu YX, Sunwoo J, and Levy R
- Published
- 2019
- Full Text
- View/download PDF
15. Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer.
- Author
-
Kohrt HE, Houot R, Weiskopf K, Goldstein MJ, Scheeren F, Czerwinski D, Colevas AD, Weng WK, Clarke MF, Carlson RW, Stockdale FE, Mollick JA, Chen L, and Levy R
- Published
- 2019
- Full Text
- View/download PDF
16. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy.
- Author
-
Barkal AA, Weiskopf K, Kao KS, Gordon SR, Rosental B, Yiu YY, George BM, Markovic M, Ring NG, Tsai JM, McKenna KM, Ho PY, Cheng RZ, Chen JY, Barkal LJ, Ring AM, Weissman IL, and Maute RL
- Subjects
- Animals, Cell Line, Tumor, Histocompatibility Antigens Class I metabolism, Humans, Immunotherapy methods, Leukocyte Immunoglobulin-like Receptor B1 metabolism, Macrophages metabolism, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, Knockout, Mice, SCID, Neoplasms metabolism, Neoplasms therapy, Neoplasms, Experimental immunology, Neoplasms, Experimental metabolism, Neoplasms, Experimental therapy, Histocompatibility Antigens Class I immunology, Leukocyte Immunoglobulin-like Receptor B1 immunology, Macrophages immunology, Neoplasms immunology, Phagocytosis immunology
- Abstract
Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component β
2 -microglobulin (β2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.- Published
- 2018
- Full Text
- View/download PDF
17. Anti-SIRPα antibody immunotherapy enhances neutrophil and macrophage antitumor activity.
- Author
-
Ring NG, Herndler-Brandstetter D, Weiskopf K, Shan L, Volkmer JP, George BM, Lietzenmayer M, McKenna KM, Naik TJ, McCarty A, Zheng Y, Ring AM, Flavell RA, and Weissman IL
- Subjects
- Animals, Antigens, Differentiation genetics, Burkitt Lymphoma genetics, Burkitt Lymphoma immunology, Burkitt Lymphoma pathology, CD27 Ligand genetics, CD27 Ligand immunology, CD47 Antigen genetics, CD47 Antigen immunology, Cell Line, Tumor, Combined Modality Therapy methods, Gene Expression, Gene Knock-In Techniques, Humans, Immunotherapy methods, Macrophages cytology, Macrophages drug effects, Macrophages immunology, Mice, Mice, Transgenic, Neutrophils cytology, Neutrophils drug effects, Neutrophils immunology, Protein Binding, Receptors, Immunologic genetics, Transgenes, Xenograft Model Antitumor Assays, Antibodies, Bispecific pharmacology, Antibodies, Monoclonal pharmacology, Antibodies, Neoplasm pharmacology, Antigens, Differentiation immunology, Burkitt Lymphoma therapy, Phagocytosis drug effects, Receptors, Immunologic immunology
- Abstract
Cancer immunotherapy has emerged as a promising therapeutic intervention. However, complete and durable responses are only seen in a fraction of patients who have cancer. A key factor that limits therapeutic success is the infiltration of tumors by cells of the myeloid lineage. The inhibitory receptor signal regulatory protein-α (SIRPα) is a myeloid-specific immune checkpoint that engages the "don't eat me" signal CD47 expressed on tumors and normal tissues. We therefore developed the monoclonal antibody KWAR23, which binds human SIRPα with high affinity and disrupts its binding to CD47. Administered by itself, KWAR23 is inert, but given in combination with tumor-opsonizing monoclonal antibodies, KWAR23 greatly augments myeloid cell-dependent killing of a collection of hematopoietic and nonhematopoietic human tumor-derived cell lines. Following KWAR23 antibody treatment in a human SIRPA knockin mouse model, both neutrophils and macrophages infiltrate a human Burkitt's lymphoma xenograft and inhibit tumor growth, generating complete responses in the majority of treated animals. We further demonstrate that a bispecific anti-CD70/SIRPα antibody outperforms individually delivered antibodies in specific types of cancers. These studies demonstrate that SIRPα blockade induces potent antitumor activity by targeting multiple myeloid cell subsets that frequently infiltrate tumors. Thus, KWAR23 represents a promising candidate for combination therapy., Competing Interests: Conflict of interest statement: K.W., A.M.R., and I.L.W. are shareholders of Forty Seven, Inc., and have filed a patent application that describes the human anti-SIRPα antibody KWAR23. I.L.W. is co-inventor of the patent, and co-founder and director of the company that has licensed the antibody.
- Published
- 2017
- Full Text
- View/download PDF
18. Delivery of monocyte lineage cells in a biomimetic scaffold enhances tissue repair.
- Author
-
Hu MS, Walmsley GG, Barnes LA, Weiskopf K, Rennert RC, Duscher D, Januszyk M, Maan ZN, Hong WX, Cheung AT, Leavitt T, Marshall CD, Ransom RC, Malhotra S, Moore AL, Rajadas J, Lorenz HP, Weissman IL, Gurtner GC, and Longaker MT
- Subjects
- Acute-Phase Proteins metabolism, Animals, Biomimetics, Cell Differentiation physiology, Diabetes Mellitus, Experimental immunology, Immunocompromised Host, Mice, Inbred Strains, Monocytes transplantation, Skin injuries, Skin Physiological Phenomena immunology, Wound Healing immunology, Diabetes Mellitus, Experimental physiopathology, Macrophages transplantation, Tissue Scaffolds, Wound Healing physiology
- Abstract
The monocyte lineage is essential to normal wound healing. Macrophage inhibition or knockout in mice results in impaired wound healing through reduced neovascularization, granulation tissue formation, and reepithelialization. Numerous studies have either depleted macrophages or reduced their activity in the context of wound healing. Here, we demonstrate that by increasing the number of macrophages or monocytes in the wound site above physiologic levels via pullulan-collagen composite dermal hydrogel scaffold delivery, the rate of wound healing can be significantly accelerated in both wild-type and diabetic mice, with no adverse effect on the quality of repair. Macrophages transplanted onto wounds differentiate into M1 and M2 phenotypes of different proportions at various time points, ultimately increasing angiogenesis. Given that monocytes can be readily isolated from peripheral blood without in vitro manipulation, these findings hold promise for translational medicine aimed at accelerating wound healing across a broad spectrum of diseases.
- Published
- 2017
- Full Text
- View/download PDF
19. Cancer immunotherapy targeting the CD47/SIRPα axis.
- Author
-
Weiskopf K
- Subjects
- Animals, Antibodies, Neoplasm immunology, Antigen Presentation drug effects, Antigen Presentation immunology, Antigens, Differentiation immunology, Humans, Macrophages immunology, Mice, Molecular Targeted Therapy, Myeloid Cells drug effects, Myeloid Cells immunology, Neoplasms immunology, Phagocytosis immunology, Receptors, Immunologic immunology, Antibodies, Neoplasm drug effects, CD47 Antigen immunology, Immunotherapy methods, Macrophages drug effects, Neoplasms drug therapy, Phagocytosis drug effects, Receptors, Immunologic antagonists & inhibitors
- Abstract
The success of cancer immunotherapy has generated tremendous interest in identifying new immunotherapeutic targets. To date, the majority of therapies have focussed on stimulating the adaptive immune system to attack cancer, including agents targeting CTLA-4 and the PD-1/PD-L1 axis. However, macrophages and other myeloid immune cells offer much promise as effectors of cancer immunotherapy. The CD47/signal regulatory protein alpha (SIRPα) axis is a critical regulator of myeloid cell activation and serves a broader role as a myeloid-specific immune checkpoint. CD47 is highly expressed on many different types of cancer, and it transduces inhibitory signals through SIRPα on macrophages and other myeloid cells. In a diverse range of preclinical models, therapies that block the CD47/SIRPα axis stimulate phagocytosis of cancer cells in vitro and anti-tumour immune responses in vivo. A number of therapeutics that target the CD47/SIRPα axis are under preclinical and clinical investigation. These include anti-CD47 antibodies, engineered receptor decoys, anti-SIRPα antibodies and bispecific agents. These therapeutics differ in their pharmacodynamic, pharmacokinetic and toxicological properties. Clinical trials are underway for both solid and haematologic malignancies using anti-CD47 antibodies and recombinant SIRPα proteins. Since the CD47/SIRPα axis also limits the efficacy of tumour-opsonising antibodies, additional trials will examine their potential synergy with agents such as rituximab, cetuximab and trastuzumab. Phagocytosis in response to CD47/SIRPα-blocking agents results in antigen uptake and presentation, thereby linking the innate and adaptive immune systems. CD47/SIRPα blocking therapies may therefore synergise with immune checkpoint inhibitors that target the adaptive immune system. As a critical regulator of macrophage phagocytosis and activation, the potential applications of CD47/SIRPα blocking therapies extend beyond human cancer. They may be useful for the treatment of infectious disease, conditioning for stem cell transplant, and many other clinical indications., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
20. Eradication of Canine Diffuse Large B-Cell Lymphoma in a Murine Xenograft Model with CD47 Blockade and Anti-CD20.
- Author
-
Weiskopf K, Anderson KL, Ito D, Schnorr PJ, Tomiyasu H, Ring AM, Bloink K, Efe J, Rue S, Lowery D, Barkal A, Prohaska S, McKenna KM, Cornax I, O'Brien TD, O'Sullivan MG, Weissman IL, and Modiano JF
- Subjects
- Animals, Cell Line, Tumor, Dogs, Female, Immunoglobulin G therapeutic use, Lymphoma, Large B-Cell, Diffuse immunology, Lymphoma, Large B-Cell, Diffuse veterinary, Macrophages immunology, Male, Mice, Phagocytosis, Xenograft Model Antitumor Assays, Antigens, CD20 immunology, CD47 Antigen immunology, Immunotherapy, Lymphoma, Large B-Cell, Diffuse therapy
- Abstract
Cancer immunotherapies hold much promise, but their potential in veterinary settings has not yet been fully appreciated. Canine lymphomas are among the most common tumors of dogs and bear remarkable similarity to human disease. In this study, we examined the combination of CD47 blockade with anti-CD20 passive immunotherapy for canine lymphoma. The CD47/SIRPα axis is an immune checkpoint that regulates macrophage activation. In humans, CD47 is expressed on cancer cells and enables evasion from phagocytosis. CD47-blocking therapies are now under investigation in clinical trials for a variety of human cancers. We found the canine CD47/SIRPα axis to be conserved biochemically and functionally. We identified high-affinity SIRPα variants that antagonize canine CD47 and stimulate phagocytosis of canine cancer cells in vitro When tested as Fc fusion proteins, these therapeutic agents exhibited single-agent efficacy in a mouse xenograft model of canine lymphoma. As robust synergy between CD47 blockade and tumor-specific antibodies has been demonstrated for human cancer, we evaluated the combination of CD47 blockade with 1E4-cIgGB, a canine-specific antibody to CD20. 1E4-cIgGB could elicit a therapeutic response against canine lymphoma in vivo as a single agent. However, augmented responses were observed when combined with CD47-blocking therapies, resulting in synergy in vitro and in vivo and eliciting cures in 100% of mice bearing canine lymphoma. Our findings support further testing of CD47-blocking therapies alone and in combination with CD20 antibodies in the veterinary setting. Cancer Immunol Res; 4(12); 1072-87. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
21. SIRPα-Antibody Fusion Proteins Selectively Bind and Eliminate Dual Antigen-Expressing Tumor Cells.
- Author
-
Piccione EC, Juarez S, Tseng S, Liu J, Stafford M, Narayanan C, Wang L, Weiskopf K, and Majeti R
- Subjects
- Animals, Antibodies, Bispecific immunology, Antigens, Differentiation chemistry, Antigens, Differentiation immunology, Antineoplastic Agents chemistry, CD47 Antigen immunology, Cell Line, Tumor, Humans, Macaca fascicularis, Male, Mice, Phagocytosis, Receptors, Immunologic chemistry, Receptors, Immunologic immunology, Rituximab chemistry, Antibodies, Bispecific pharmacology, Antineoplastic Agents pharmacology, CD47 Antigen antagonists & inhibitors, Receptors, Immunologic antagonists & inhibitors, Rituximab pharmacology
- Abstract
Purpose: CD47 is highly expressed on a variety of tumor cells. The interaction of CD47 with signal regulatory protein alpha (SIRPα), a protein on phagocytic cells, transmits a "don't eat me" signal that negatively regulates phagocytosis. CD47-SIRPα antagonists enable phagocytosis by disrupting the inhibitory signal and can synergize with Fc-mediated pro-phagocytic signals for potent elimination of tumor cells. A potential limitation of therapeutic CD47-SIRPα antagonists is that expression of CD47 on normal cells may create sites of toxicity or an "antigen sink." To overcome these limitations and address selective tumor targeting, we developed SIRPabodies to improve the therapeutic benefits of CD47-SIRPα blockade specifically toward tumor., Experimental Design: SIRPabodies were generated by grafting the wild-type SIRPα either to the N-terminus or to the C-terminus of the heavy chain of rituximab. Selective tumor binding was tested using CFSE-labeled human primary CLL cells in the presence of 20-fold excess of human RBCs. NSG mice were transplanted with Raji-luciferase cells and were assigned to controls versus SIRPabody treatment. Cynomolgus nonhuman primates were administered a single intravenous infusion of SIRPabody at 3, 10, or 30 mg/kg., Results: SIRPabodies selectively bound to dual antigen-expressing tumor cells in the presence of a large antigen sink. SIRPabody reduced tumor burden and extended survival in mouse xenograft lymphoma models. SIRPabody caused no significant toxicity in nonhuman primates., Conclusions: These findings establish SIRPabodies as a promising approach to deliver the therapeutic benefit of CD47-SIRPα blockade specifically toward tumor cells. SIRPabodies may be applied to additional cancer types by grafting SIRPα onto other tumor-specific therapeutic antibodies. Clin Cancer Res; 22(20); 5109-19. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
22. Myeloid Cell Origins, Differentiation, and Clinical Implications.
- Author
-
Weiskopf K, Schnorr PJ, Pang WW, Chao MP, Chhabra A, Seita J, Feng M, and Weissman IL
- Subjects
- Animals, Cell Differentiation physiology, Cell Lineage, Humans, Hematopoietic Stem Cells cytology, Myeloid Cells cytology
- Abstract
The hematopoietic stem cell (HSC) is a multipotent stem cell that resides in the bone marrow and has the ability to form all of the cells of the blood and immune system. Since its first purification in 1988, additional studies have refined the phenotype and functionality of HSCs and characterized all of their downstream progeny. The hematopoietic lineage is divided into two main branches: the myeloid and lymphoid arms. The myeloid arm is characterized by the common myeloid progenitor and all of its resulting cell types. The stages of hematopoiesis have been defined in both mice and humans. During embryological development, the earliest hematopoiesis takes place in yolk sac blood islands and then migrates to the fetal liver and hematopoietic organs. Some adult myeloid populations develop directly from yolk sac progenitors without apparent bone marrow intermediates, such as tissue-resident macrophages. Hematopoiesis also changes over time, with a bias of the dominating HSCs toward myeloid development as animals age. Defects in myelopoiesis contribute to many hematologic disorders, and some of these can be overcome with therapies that target the aberrant stage of development. Furthermore, insights into myeloid development have informed us of mechanisms of programmed cell removal. The CD47/SIRPα axis, a myeloid-specific immune checkpoint, limits macrophage removal of HSCs but can be exploited by hematologic and solid malignancies. Therapeutics targeting CD47 represent a new strategy for treating cancer. Overall, an understanding of hematopoiesis and myeloid cell development has implications for regenerative medicine, hematopoietic cell transplantation, malignancy, and many other diseases.
- Published
- 2016
- Full Text
- View/download PDF
23. Salmonella Infection Enhances Erythropoietin Production by the Kidney and Liver, Which Correlates with Elevated Bacterial Burdens.
- Author
-
Li LX, Benoun JM, Weiskopf K, Garcia KC, and McSorley SJ
- Subjects
- Anemia blood, Animals, Bacterial Load, Disease Models, Animal, Erythropoiesis physiology, Flow Cytometry, Mice, Mice, Inbred C57BL, RNA, Messenger metabolism, Receptors, Erythropoietin metabolism, Receptors, Immunologic metabolism, Spleen metabolism, Erythropoietin metabolism, Kidney metabolism, Liver metabolism, Salmonella Infections metabolism, Salmonella Infections microbiology, Salmonella typhi
- Abstract
Salmonella infection profoundly affects host erythroid development, but the mechanisms responsible for this effect remain poorly understood. We monitored the impact of Salmonella infection on erythroid development and found that systemic infection induced anemia, splenomegaly, elevated erythropoietin (EPO) levels, and extramedullary erythropoiesis in a process independent of Salmonella pathogenicity island 2 (SPI2) or flagellin. The circulating EPO level was also constitutively higher in mice lacking the expression of signal-regulatory protein α (SIRPα). The expression level of EPO mRNA was elevated in the kidney and liver but not increased in the spleens of infected mice despite the presence of extramedullary erythropoiesis in this tissue. In contrast to data from a previous report, mice lacking EPO receptor (EPOR) expression on nonerythroid cells (EPOR rescued) had bacterial loads similar to those of wild-type mice following Salmonella infection. Indeed, treatment to reduce splenic erythroblasts and mature red blood cells correlated with elevated bacterial burdens, implying that extramedullary erythropoiesis benefits the host. Together, these findings emphasize the profound effect of Salmonella infection on erythroid development and suggest that the modulation of erythroid development has both positive and negative consequences for host immunity., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
24. Hematopoietic stem cell transplantation in immunocompetent hosts without radiation or chemotherapy.
- Author
-
Chhabra A, Ring AM, Weiskopf K, Schnorr PJ, Gordon S, Le AC, Kwon HS, Ring NG, Volkmer J, Ho PY, Tseng S, Weissman IL, and Shizuru JA
- Subjects
- Animals, CD47 Antigen antagonists & inhibitors, CD47 Antigen metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Erythrocytes metabolism, Flow Cytometry, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells physiology, Humans, Mice, Mice, Mutant Strains, Receptors, Fc genetics, Receptors, Fc metabolism, Hematopoietic Stem Cell Transplantation methods, Immunotherapy methods
- Abstract
Hematopoietic stem cell (HSC) transplantation can cure diverse diseases of the blood system, including hematologic malignancies, anemias, and autoimmune disorders. However, patients must undergo toxic conditioning regimens that use chemotherapy and/or radiation to eliminate host HSCs and enable donor HSC engraftment. Previous studies have shown that anti-c-Kit monoclonal antibodies deplete HSCs from bone marrow niches, allowing donor HSC engraftment in immunodeficient mice. We show that host HSC clearance is dependent on Fc-mediated antibody effector functions, and enhancing effector activity through blockade of CD47, a myeloid-specific immune checkpoint, extends anti-c-Kit conditioning to fully immunocompetent mice. The combined treatment leads to elimination of >99% of host HSCs and robust multilineage blood reconstitution after HSC transplantation. This targeted conditioning regimen that uses only biologic agents has the potential to transform the practice of HSC transplantation and enable its use in a wider spectrum of patients., (Copyright © 2016, American Association for the Advancement of Science.)
- Published
- 2016
- Full Text
- View/download PDF
25. Antibody Therapy Targeting CD47 and CD271 Effectively Suppresses Melanoma Metastasis in Patient-Derived Xenografts.
- Author
-
Ngo M, Han A, Lakatos A, Sahoo D, Hachey SJ, Weiskopf K, Beck AH, Weissman IL, and Boiko AD
- Subjects
- Adapalene metabolism, CD47 Antigen metabolism, Cell Line, Tumor, Heterografts, Humans, Macrophages immunology, Melanoma pathology, Melanoma therapy, Neoplasm Metastasis, Phagocytosis physiology, Vascular Endothelial Growth Factor Receptor-1 genetics, Vascular Endothelial Growth Factor Receptor-1 metabolism, Adapalene immunology, CD47 Antigen immunology, Melanoma immunology, Melanoma metabolism
- Abstract
The high rate of metastasis and recurrence among melanoma patients indicates the existence of cells within melanoma that have the ability to both initiate metastatic programs and bypass immune recognition. Here, we identify CD47 as a regulator of melanoma tumor metastasis and immune evasion. Protein and gene expression analysis of clinical melanoma samples reveals that CD47, an anti-phagocytic signal, correlates with melanoma metastasis. Antibody-mediated blockade of CD47 coupled with targeting of CD271(+) melanoma cells strongly inhibits tumor metastasis in patient-derived xenografts. This therapeutic effect is mediated by drastic changes in the tumor and metastatic site immune microenvironments, both of whichwhich exhibit greatly increased density of differentiated macrophages and significantly fewer inflammatory monocytes, pro-metastatic macrophages (CCR2(+)/VEGFR1(+)), and neutrophils, all of which are associated with disease progression. Thus, antibody therapy that activates the innate immune response in combination with selective targeting of CD271(+) melanoma cells represents a powerful therapeutic approach against metastatic melanoma., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
26. Acute, Unilateral Breast Toxicity From Gemcitabine in the Setting of Thoracic Inlet Obstruction.
- Author
-
Weiskopf K, Creighton D, Lew T, Caswell JL, Ouyang D, Shah AT, Hofmann LV, Neal JW, and Telli ML
- Subjects
- Adenocarcinoma secondary, Antimetabolites, Antineoplastic administration & dosage, Antineoplastic Combined Chemotherapy Protocols, Breast pathology, Female, Humans, Infusions, Intravenous adverse effects, Lung Neoplasms pathology, Mastitis chemically induced, Mastitis pathology, Middle Aged, Organ Size, Adenocarcinoma drug therapy, Antimetabolites, Antineoplastic adverse effects, Breast drug effects, Lung Neoplasms drug therapy
- Published
- 2016
- Full Text
- View/download PDF
27. CD47-blocking immunotherapies stimulate macrophage-mediated destruction of small-cell lung cancer.
- Author
-
Weiskopf K, Jahchan NS, Schnorr PJ, Cristea S, Ring AM, Maute RL, Volkmer AK, Volkmer JP, Liu J, Lim JS, Yang D, Seitz G, Nguyen T, Wu D, Jude K, Guerston H, Barkal A, Trapani F, George J, Poirier JT, Gardner EE, Miles LA, de Stanchina E, Lofgren SM, Vogel H, Winslow MM, Dive C, Thomas RK, Rudin CM, van de Rijn M, Majeti R, Garcia KC, Weissman IL, and Sage J
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, CD56 Antigen metabolism, Cell Line, Tumor, Cytokines metabolism, Green Fluorescent Proteins metabolism, Humans, Lung Neoplasms immunology, Mice, Phagocytosis, Receptors, Immunologic metabolism, Signal Transduction, Small Cell Lung Carcinoma immunology, CD47 Antigen metabolism, Immunotherapy methods, Lung Neoplasms therapy, Macrophages immunology, Small Cell Lung Carcinoma therapy
- Abstract
Small-cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer with limited treatment options. CD47 is a cell-surface molecule that promotes immune evasion by engaging signal-regulatory protein alpha (SIRPα), which serves as an inhibitory receptor on macrophages. Here, we found that CD47 is highly expressed on the surface of human SCLC cells; therefore, we investigated CD47-blocking immunotherapies as a potential approach for SCLC treatment. Disruption of the interaction of CD47 with SIRPα using anti-CD47 antibodies induced macrophage-mediated phagocytosis of human SCLC patient cells in culture. In a murine model, administration of CD47-blocking antibodies or targeted inactivation of the Cd47 gene markedly inhibited SCLC tumor growth. Furthermore, using comprehensive antibody arrays, we identified several possible therapeutic targets on the surface of SCLC cells. Antibodies to these targets, including CD56/neural cell adhesion molecule (NCAM), promoted phagocytosis in human SCLC cell lines that was enhanced when combined with CD47-blocking therapies. In light of recent clinical trials for CD47-blocking therapies in cancer treatment, these findings identify disruption of the CD47/SIRPα axis as a potential immunotherapeutic strategy for SCLC. This approach could enable personalized immunotherapeutic regimens in patients with SCLC and other cancers.
- Published
- 2016
- Full Text
- View/download PDF
28. Targeting lymphoma with precision using semisynthetic anti-idiotype peptibodies.
- Author
-
Torchia J, Weiskopf K, and Levy R
- Subjects
- Animals, Antibodies, Anti-Idiotypic immunology, Cancer Vaccines immunology, Drug Design, Female, Humans, Lymphoma pathology, Mice, Middle Aged, Molecular Targeted Therapy methods, Recombinant Fusion Proteins administration & dosage, Treatment Outcome, Tumor Cells, Cultured, Vaccines, Synthetic immunology, Antibodies, Anti-Idiotypic administration & dosage, Cancer Vaccines administration & dosage, Lymphoma immunology, Lymphoma therapy, Protein Engineering methods, Vaccines, Synthetic administration & dosage
- Abstract
B-cell lymphomas express a functionally active and truly tumor-specific cell-surface product, the variable region of the B-cell receptor (BCR), otherwise known as idiotype. The tumor idiotype differs, however, from patient to patient, making it a technical challenge to exploit for therapy. We have developed a method of targeting idiotype by using a semisynthetic personalized therapeutic that is more practical to produce on a patient-by-patient basis than monoclonal antibodies. In this method, a small peptide with affinity for a tumor idiotype is identified by screening a library, chemically synthesized, and then affixed to the amino terminus of a premade IgG Fc protein. We demonstrate that the resultant semisynthetic anti-idiotype peptibodies kill tumor cells in vitro with specificity, trigger tumor cell phagocytosis by macrophages, and efficiently clear human lymphoma in a murine xenograft model. This method could be used to target tumor with true precision on a personalized basis.
- Published
- 2016
- Full Text
- View/download PDF
29. Identification of tumorigenic cells and therapeutic targets in pancreatic neuroendocrine tumors.
- Author
-
Krampitz GW, George BM, Willingham SB, Volkmer JP, Weiskopf K, Jahchan N, Newman AM, Sahoo D, Zemek AJ, Yanovsky RL, Nguyen JK, Schnorr PJ, Mazur PK, Sage J, Longacre TA, Visser BC, Poultsides GA, Norton JA, and Weissman IL
- Subjects
- Aldehyde Dehydrogenase 1 Family, Animals, CD47 Antigen immunology, Female, Humans, Isoenzymes immunology, Male, Mice, Inbred NOD, Mice, SCID, Neoplasm Metastasis, Neoplasm Proteins immunology, Proto-Oncogene Mas, Retinal Dehydrogenase immunology, Thy-1 Antigens immunology, Xenograft Model Antitumor Assays, Neuroendocrine Tumors immunology, Neuroendocrine Tumors pathology, Neuroendocrine Tumors therapy, Pancreatic Neoplasms immunology, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy, Tumor Escape
- Abstract
Pancreatic neuroendocrine tumors (PanNETs) are a type of pancreatic cancer with limited therapeutic options. Consequently, most patients with advanced disease die from tumor progression. Current evidence indicates that a subset of cancer cells is responsible for tumor development, metastasis, and recurrence, and targeting these tumor-initiating cells is necessary to eradicate tumors. However, tumor-initiating cells and the biological processes that promote pathogenesis remain largely uncharacterized in PanNETs. Here we profile primary and metastatic tumors from an index patient and demonstrate that MET proto-oncogene activation is important for tumor growth in PanNET xenograft models. We identify a highly tumorigenic cell population within several independent surgically acquired PanNETs characterized by increased cell-surface protein CD90 expression and aldehyde dehydrogenase A1 (ALDHA1) activity, and provide in vitro and in vivo evidence for their stem-like properties. We performed proteomic profiling of 332 antigens in two cell lines and four primary tumors, and showed that CD47, a cell-surface protein that acts as a "don't eat me" signal co-opted by cancers to evade innate immune surveillance, is ubiquitously expressed. Moreover, CD47 coexpresses with MET and is enriched in CD90(hi)cells. Furthermore, blocking CD47 signaling promotes engulfment of tumor cells by macrophages in vitro and inhibits xenograft tumor growth, prevents metastases, and prolongs survival in vivo.
- Published
- 2016
- Full Text
- View/download PDF
30. Interleukin-2 activity can be fine tuned with engineered receptor signaling clamps.
- Author
-
Mitra S, Ring AM, Amarnath S, Spangler JB, Li P, Ju W, Fischer S, Oh J, Spolski R, Weiskopf K, Kohrt H, Foley JE, Rajagopalan S, Long EO, Fowler DH, Waldmann TA, Garcia KC, and Leonard WJ
- Subjects
- Animals, Cell Line, Cell Proliferation, Female, Gene Expression Regulation, Graft vs Host Disease, Humans, Interleukin-2 chemistry, Interleukin-2 genetics, Leukemia-Lymphoma, Adult T-Cell immunology, Leukemia-Lymphoma, Adult T-Cell physiopathology, Mice, Mice, Inbred C57BL, Models, Molecular, Mutation, Protein Binding, Protein Structure, Tertiary, Receptors, Interleukin-2 chemistry, STAT5 Transcription Factor metabolism, Survival Analysis, Interleukin-2 antagonists & inhibitors, Protein Engineering, Receptors, Interleukin-2 metabolism, Signal Transduction immunology
- Abstract
Interleukin-2 (IL-2) regulates lymphocyte function by signaling through heterodimerization of the IL-2Rβ and γc receptor subunits. IL-2 is of considerable therapeutic interest, but harnessing its actions in a controllable manner remains a challenge. Previously, we have engineered an IL-2 "superkine" with enhanced affinity for IL-2Rβ. Here, we describe next-generation IL-2 variants that function as "receptor signaling clamps." They retained high affinity for IL-2Rβ, inhibiting binding of endogenous IL-2, but their interaction with γc was weakened, attenuating IL-2Rβ-γc heterodimerization. These IL-2 analogs acted as partial agonists and differentially affected lymphocytes poised at distinct activation thresholds. Moreover, one variant, H9-RETR, antagonized IL-2 and IL-15 better than blocking antibodies against IL-2Rα or IL-2Rβ. Furthermore, this mutein prolonged survival in a model of graft-versus-host disease and blocked spontaneous proliferation of smoldering adult T cell leukemia (ATL) T cells. This receptor-clamping approach might be a general mechanism-based strategy for engineering cytokine partial agonists for therapeutic immunomodulation., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
31. "Velcro" engineering of high affinity CD47 ectodomain as signal regulatory protein α (SIRPα) antagonists that enhance antibody-dependent cellular phagocytosis.
- Author
-
Ho CC, Guo N, Sockolosky JT, Ring AM, Weiskopf K, Özkan E, Mori Y, Weissman IL, and Garcia KC
- Subjects
- Animals, Antibodies, Antigens, Differentiation genetics, Antigens, Differentiation metabolism, Cell Line, Tumor, Humans, Immunotherapy, Macrophages pathology, Mice, Neoplasms genetics, Neoplasms metabolism, Protein Binding, Protein Engineering, Protein Structure, Tertiary, Receptors, Immunologic genetics, Receptors, Immunologic metabolism, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, CD47 Antigen chemistry, CD47 Antigen genetics, CD47 Antigen pharmacology, Macrophages metabolism, Neoplasms drug therapy, Phagocytosis drug effects, Receptors, Immunologic antagonists & inhibitors
- Abstract
CD47 is a cell surface protein that transmits an anti-phagocytic signal, known as the "don't-eat-me" signal, to macrophages upon engaging its receptor signal regulatory protein α (SIRPα). Molecules that antagonize the CD47-SIRPα interaction by binding to CD47, such as anti-CD47 antibodies and the engineered SIRPα variant CV1, have been shown to facilitate macrophage-mediated anti-tumor responses. However, these strategies targeting CD47 are handicapped by large antigen sinks in vivo and indiscriminate cell binding due to ubiquitous expression of CD47. These factors reduce bioavailability and increase the risk of toxicity. Here, we present an alternative strategy to antagonize the CD47-SIRPα pathway by engineering high affinity CD47 variants that target SIRPα, which has restricted tissue expression. CD47 proved to be refractive to conventional affinity maturation techniques targeting its binding interface with SIRPα. Therefore, we developed a novel engineering approach, whereby we augmented the existing contact interface via N-terminal peptide extension, coined "Velcro" engineering. The high affinity variant (Velcro-CD47) bound to the two most prominent human SIRPα alleles with greatly increased affinity relative to wild-type CD47 and potently antagonized CD47 binding to SIRPα on human macrophages. Velcro-CD47 synergizes with tumor-specific monoclonal antibodies to enhance macrophage phagocytosis of tumor cells in vitro, with similar potency as CV1. Finally, Velcro-CD47 interacts specifically with a subset of myeloid-derived cells in human blood, whereas CV1 binds all myeloid, lymphoid, and erythroid populations interrogated. This is consistent with the restricted expression of SIRPα compared with CD47. Herein, we have demonstrated that "Velcro" engineering is a powerful protein-engineering tool with potential applications to other systems and that Velcro-CD47 could be an alternative adjuvant to CD47-targeting agents for cancer immunotherapy., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
32. A bispecific antibody targeting CD47 and CD20 selectively binds and eliminates dual antigen expressing lymphoma cells.
- Author
-
Piccione EC, Juarez S, Liu J, Tseng S, Ryan CE, Narayanan C, Wang L, Weiskopf K, and Majeti R
- Subjects
- Animals, Antibodies, Bispecific immunology, Antibodies, Monoclonal immunology, Antibody Affinity, Humans, Immunoglobulin G immunology, Immunoglobulin G pharmacology, Mice, Polymerase Chain Reaction, Surface Plasmon Resonance, Xenograft Model Antitumor Assays, Antibodies, Bispecific pharmacology, Antibodies, Monoclonal pharmacology, Antigens, CD20 immunology, CD47 Antigen immunology, Lymphoma, Non-Hodgkin immunology
- Abstract
Agents that block the anti-phagocytic signal CD47 can synergize with pro-phagocytic anti-tumor antigen antibodies to potently eliminate tumors. While CD47 is overexpressed on cancer cells, its expression in many normal tissues may create an 'antigen sink' that could minimize the therapeutic efficacy of CD47 blocking agents. Here, we report development of bispecific antibodies (BsAbs) that co-target CD47 and CD20, a therapeutic target for non-Hodgkin lymphoma (NHL), that have reduced affinity for CD47 relative to the parental antibody, but retain strong binding to CD20. These characteristics facilitate selective binding of BsAbs to tumor cells, leading to phagocytosis. Treatment of human NHL-engrafted mice with BsAbs reduced lymphoma burden and extended survival while recapitulating the synergistic efficacy of anti-CD47 and anti-CD20 combination therapy. These findings serve as proof of principle for BsAb targeting of CD47 with tumor-associated antigens as a viable strategy to induce selective phagocytosis of tumor cells and recapitulate the synergy of combination antibody therapy. This approach may be broadly applied to cancer to add a CD47 blocking component to existing antibody therapies.
- Published
- 2015
- Full Text
- View/download PDF
33. Macrophages are critical effectors of antibody therapies for cancer.
- Author
-
Weiskopf K and Weissman IL
- Subjects
- Animals, Humans, Antibodies, Neoplasm immunology, Antibodies, Neoplasm therapeutic use, Macrophages immunology, Neoplasms drug therapy, Neoplasms immunology, Phagocytosis drug effects, Receptors, IgG immunology
- Abstract
Macrophages are innate immune cells that derive from circulating monocytes, reside in all tissues, and participate in many states of pathology. Macrophages play a dichotomous role in cancer, where they promote tumor growth but also serve as critical immune effectors of therapeutic antibodies. Macrophages express all classes of Fcγ receptors, and they have immense potential to destroy tumors via the process of antibody-dependent phagocytosis. A number of studies have demonstrated that macrophage phagocytosis is a major mechanism of action of many antibodies approved to treat cancer. Consequently, a number of approaches to augment macrophage responses to therapeutic antibodies are under investigation, including the exploration of new targets and development of antibodies with enhanced functions. For example, the interaction of CD47 with signal-regulatory protein α (SIRPα) serves as a myeloid-specific immune checkpoint that limits the response of macrophages to antibody therapies, and CD47-blocking agents overcome this barrier to augment phagocytosis. The response of macrophages to antibody therapies can also be enhanced with engineered Fc variants, bispecific antibodies, or antibody-drug conjugates. Macrophages have demonstrated success as effectors of cancer immunotherapy, and further investigation will unlock their full potential for the benefit of patients.
- Published
- 2015
- Full Text
- View/download PDF
34. Q&A: Michael Bishop. Free thinker.
- Author
-
Bishop M and Weiskopf K
- Subjects
- Animals, Biomedical Research, California, Computational Biology, Drug Discovery organization & administration, Drug Discovery trends, Drug Industry, Drug Resistance, Neoplasm genetics, Female, History, 20th Century, Nobel Prize, RNA-Directed DNA Polymerase history, Retroviridae enzymology, Smoking adverse effects, Neoplasms etiology, Neoplasms genetics, Neoplasms metabolism
- Published
- 2014
- Full Text
- View/download PDF
35. Targeting CD137 enhances the efficacy of cetuximab.
- Author
-
Kohrt HE, Colevas AD, Houot R, Weiskopf K, Goldstein MJ, Lund P, Mueller A, Sagiv-Barfi I, Marabelle A, Lira R, Troutner E, Richards L, Rajapaska A, Hebb J, Chester C, Waller E, Ostashko A, Weng WK, Chen L, Czerwinski D, Fu YX, Sunwoo J, and Levy R
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibody-Dependent Cell Cytotoxicity, Antineoplastic Agents administration & dosage, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Cetuximab, Colorectal Neoplasms genetics, Colorectal Neoplasms immunology, Colorectal Neoplasms therapy, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Female, Head and Neck Neoplasms genetics, Head and Neck Neoplasms immunology, Head and Neck Neoplasms therapy, Humans, Immunotherapy, Adoptive, Killer Cells, Natural immunology, Mice, Mice, Inbred BALB C, Mice, Nude, Mutation, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins p21(ras), Tumor Necrosis Factor Receptor Superfamily, Member 9 metabolism, ras Proteins genetics, Antibodies, Monoclonal, Humanized administration & dosage, Tumor Necrosis Factor Receptor Superfamily, Member 9 antagonists & inhibitors
- Abstract
Treatment with cetuximab, an EGFR-targeting IgG1 mAb, results in beneficial, yet limited, clinical improvement for patients with head and neck (HN) cancer as well as colorectal cancer (CRC) patients with WT KRAS tumors. Antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells contributes to the efficacy of cetuximab. The costimulatory molecule CD137 (4-1BB) is expressed following NK and memory T cell activation. We found that isolated human NK cells substantially increased expression of CD137 when exposed to cetuximab-coated, EGFR-expressing HN and CRC cell lines. Furthermore, activation of CD137 with an agonistic mAb enhanced NK cell degranulation and cytotoxicity. In multiple murine xenograft models, including EGFR-expressing cancer cells, HN cells, and KRAS-WT and KRAS-mutant CRC, combined cetuximab and anti-CD137 mAb administration was synergistic and led to complete tumor resolution and prolonged survival, which was dependent on the presence of NK cells. In patients receiving cetuximab, the level of CD137 on circulating and intratumoral NK cells was dependent on postcetuximab time and host FcyRIIIa polymorphism. Interestingly, the increase in CD137-expressing NK cells directly correlated to an increase in EGFR-specific CD8+ T cells. These results support development of a sequential antibody approach against EGFR-expressing malignancies that first targets the tumor and then the host immune system.
- Published
- 2014
- Full Text
- View/download PDF
36. Improving macrophage responses to therapeutic antibodies by molecular engineering of SIRPα variants.
- Author
-
Weiskopf K, Ring AM, Schnorr PJ, Volkmer JP, Volkmer AK, Weissman IL, and Garcia KC
- Abstract
CD47 transduces inhibitory signals through signal-regulatory protein α (SIRPα), a plasma membrane receptor expressed by macrophages. Many cancers upregulate CD47 to evade immunosurveillance. We have recently engineered SIRPα variants that potently antagonize CD47 for use as anticancer immunotherapeutics. These high-affinity SIRPα variants synergize with antineoplastic antibodies by lowering the threshold for macrophage-mediated destruction of malignant cells.
- Published
- 2013
- Full Text
- View/download PDF
37. Engineered SIRPα variants as immunotherapeutic adjuvants to anticancer antibodies.
- Author
-
Weiskopf K, Ring AM, Ho CC, Volkmer JP, Levin AM, Volkmer AK, Ozkan E, Fernhoff NB, van de Rijn M, Weissman IL, and Garcia KC
- Subjects
- Animals, Antibodies, Monoclonal, Murine-Derived therapeutic use, Antigens, Differentiation chemistry, Antigens, Differentiation genetics, Cell Line, Tumor, Directed Molecular Evolution, Humans, Immunotherapy, Macrophage Activation, Mice, Neoplasms immunology, Phagocytosis, Receptors, Immunologic chemistry, Receptors, Immunologic genetics, Rituximab, Adjuvants, Immunologic, Antibodies, Monoclonal therapeutic use, Antibodies, Neoplasm therapeutic use, Antigens, Differentiation therapeutic use, CD47 Antigen immunology, Neoplasms therapy, Receptors, Immunologic therapeutic use
- Abstract
CD47 is an antiphagocytic signal that cancer cells employ to inhibit macrophage-mediated destruction. Here, we modified the binding domain of human SIRPα, the receptor for CD47, for use as a CD47 antagonist. We engineered high-affinity SIRPα variants with about a 50,000-fold increased affinity for human CD47 relative to wild-type SIRPα. As high-affinity SIRPα monomers, they potently antagonized CD47 on cancer cells but did not induce macrophage phagocytosis on their own. Instead, they exhibited remarkable synergy with all tumor-specific monoclonal antibodies tested by increasing phagocytosis in vitro and enhancing antitumor responses in vivo. This "one-two punch" directs immune responses against tumor cells while lowering the threshold for macrophage activation, thereby providing a universal method for augmenting the efficacy of therapeutic anticancer antibodies.
- Published
- 2013
- Full Text
- View/download PDF
38. Anti-CD47 antibody-mediated phagocytosis of cancer by macrophages primes an effective antitumor T-cell response.
- Author
-
Tseng D, Volkmer JP, Willingham SB, Contreras-Trujillo H, Fathman JW, Fernhoff NB, Seita J, Inlay MA, Weiskopf K, Miyanishi M, and Weissman IL
- Subjects
- Animals, Antibodies, Blocking therapeutic use, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Colonic Neoplasms pathology, Cytotoxicity, Immunologic genetics, Down-Regulation genetics, Down-Regulation immunology, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phagocytosis immunology, Up-Regulation genetics, Up-Regulation immunology, Xenograft Model Antitumor Assays, Antibodies, Monoclonal therapeutic use, CD47 Antigen immunology, Colonic Neoplasms immunology, Colonic Neoplasms therapy, Macrophages immunology, T-Lymphocytes immunology
- Abstract
Mobilization of the T-cell response against cancer has the potential to achieve long-lasting cures. However, it is not known how to harness antigen-presenting cells optimally to achieve an effective antitumor T-cell response. In this study, we show that anti-CD47 antibody-mediated phagocytosis of cancer by macrophages can initiate an antitumor T-cell immune response. Using the ovalbumin model antigen system, anti-CD47 antibody-mediated phagocytosis of cancer cells by macrophages resulted in increased priming of OT-I T cells [cluster of differentiation 8-positive (CD8(+))] but decreased priming of OT-II T cells (CD4(+)). The CD4(+) T-cell response was characterized by a reduction in forkhead box P3-positive (Foxp3(+)) regulatory T cells. Macrophages following anti-CD47-mediated phagocytosis primed CD8(+) T cells to exhibit cytotoxic function in vivo. This response protected animals from tumor challenge. We conclude that anti-CD47 antibody treatment not only enables macrophage phagocytosis of cancer but also can initiate an antitumor cytotoxic T-cell immune response.
- Published
- 2013
- Full Text
- View/download PDF
39. Use of a KIT-specific monoclonal antibody to bypass imatinib resistance in gastrointestinal stromal tumors.
- Author
-
Edris B, Willingham S, Weiskopf K, Volkmer AK, Volkmer JP, Mühlenberg T, Weissman IL, and van de Rijn M
- Abstract
Acquired resistance to imatinib is a significant problem for the clinical management of gastrointestinal stromal tumor (GIST) patients, and second-line small molecules have shown limited efficacy in this setting. We have recently demonstrated that a monoclonal antibody targeting KIT could potentially bypass imatinib resistance in preclinical models of GIST.
- Published
- 2013
- Full Text
- View/download PDF
40. Anti-KIT monoclonal antibody inhibits imatinib-resistant gastrointestinal stromal tumor growth.
- Author
-
Edris B, Willingham SB, Weiskopf K, Volkmer AK, Volkmer JP, Mühlenberg T, Montgomery KD, Contreras-Trujillo H, Czechowicz A, Fletcher JA, West RB, Weissman IL, and van de Rijn M
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Benzamides, Cell Line, Tumor, Cell Membrane drug effects, Cell Membrane metabolism, Cell Proliferation drug effects, Humans, Imatinib Mesylate, Macrophages drug effects, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phagocytosis drug effects, Piperazines pharmacology, Pyrimidines pharmacology, Xenograft Model Antitumor Assays, Antibodies, Monoclonal therapeutic use, Drug Resistance, Neoplasm drug effects, Gastrointestinal Stromal Tumors drug therapy, Gastrointestinal Stromal Tumors pathology, Piperazines therapeutic use, Proto-Oncogene Proteins c-kit immunology, Pyrimidines therapeutic use
- Abstract
Gastrointestinal stromal tumor (GIST) is the most common sarcoma of the gastrointestinal tract and arises from the interstitial cells of Cajal. It is characterized by expression of the receptor tyrosine kinase CD117 (KIT). In 70-80% of GIST cases, oncogenic mutations in KIT are present, leading to constitutive activation of the receptor, which drives the proliferation of these tumors. Treatment of GIST with imatinib, a small-molecule tyrosine kinase inhibitor, inhibits KIT-mediated signaling and initially results in disease control in 70-85% of patients with KIT-positive GIST. However, the vast majority of patients eventually develop resistance to imatinib treatment, leading to disease progression and posing a significant challenge in the clinical management of these tumors. Here, we show that an anti-KIT monoclonal antibody (mAb), SR1, is able to slow the growth of three human GIST cell lines in vitro. Importantly, these reductions in cell growth were equivalent between imatinib-resistant and imatinib-sensitive GIST cell lines. Treatment of GIST cell lines with SR1 reduces cell-surface KIT expression, suggesting that mAb-induced KIT down-regulation may be a mechanism by which SR1 inhibits GIST growth. Furthermore, we also show that SR1 treatment enhances phagocytosis of GIST cells by macrophages, indicating that treatment with SR1 may enhance immune cell-mediated tumor clearance. Finally, using two xenotransplantation models of imatinib-sensitive and imatinib-resistant GIST, we demonstrate that SR1 is able to strongly inhibit tumor growth in vivo. These results suggest that treatment with mAbs targeting KIT may represent an alternative, or complementary, approach for treating GIST.
- Published
- 2013
- Full Text
- View/download PDF
41. Flipping the script on macrophages in leiomyosarcoma.
- Author
-
Edris B, Weiskopf K, Weissman IL, and van de Rijn M
- Abstract
Macrophages promote the growth of leiomyosarcoma (LMS), a malignant soft-tissue tumor. CD47 on tumor cells binds to the macrophagic receptor signal regulatory protein α (SIRPα) and prevents phagocytosis. We showed that anti-CD47 monoclonal antibodies (mAbs) allow macrophages to engulf LMS cells and prevent tumor growth and metastases. Therefore, anti-CD47 mAbs represent a promising targeted immunotherapy for LMS.
- Published
- 2012
- Full Text
- View/download PDF
42. A nonpeptidic cathepsin S activity-based probe for noninvasive optical imaging of tumor-associated macrophages.
- Author
-
Verdoes M, Edgington LE, Scheeren FA, Leyva M, Blum G, Weiskopf K, Bachmann MH, Ellman JA, and Bogyo M
- Subjects
- Animals, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Line, Cell Line, Tumor, Cell Transformation, Neoplastic pathology, Female, Humans, Macrophages metabolism, Mice, Mice, Nude, Microscopy, Fluorescence, Prognosis, Breast Neoplasms diagnosis, Cathepsins metabolism, Fluorescent Dyes chemistry, Fluorescent Dyes metabolism, Macrophages pathology
- Abstract
Macrophage infiltration into tumors has been correlated with poor clinical outcome in multiple cancer types. Therefore, tools to image tumor-associated macrophages could be valuable for diagnosis and prognosis of cancer. Herein, we describe the synthesis and characterization of a cathepsin S-directed, quenched activity-based probe (qABP), BMV083. This probe makes use of an optimized nonpeptidic scaffold leading to enhanced in vivo properties relative to previously reported peptide-based probes. In a syngeneic breast cancer model, BMV083 provides high tumor-specific fluorescence that can be visualized using noninvasive optical imaging methods. Furthermore, analysis of probe-labeled cells demonstrates that the probe primarily targets macrophages with an M2 phenotype. Thus, BMV083 is a potential valuable in vivo reporter for tumor-associated macrophages that could greatly facilitate the future studies of macrophage function in the process of tumorigenesis., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
43. Antibody therapy targeting the CD47 protein is effective in a model of aggressive metastatic leiomyosarcoma.
- Author
-
Edris B, Weiskopf K, Volkmer AK, Volkmer JP, Willingham SB, Contreras-Trujillo H, Liu J, Majeti R, West RB, Fletcher JA, Beck AH, Weissman IL, and van de Rijn M
- Subjects
- Animals, Antibodies immunology, Cell Line, Tumor, Leiomyosarcoma pathology, Mice, Neoplasm Metastasis, Phagocytosis immunology, Antibodies therapeutic use, CD47 Antigen immunology, Disease Models, Animal, Immunotherapy, Leiomyosarcoma therapy
- Abstract
Antibodies against CD47, which block tumor cell CD47 interactions with macrophage signal regulatory protein-α, have been shown to decrease tumor size in hematological and epithelial tumor models by interfering with the protection from phagocytosis by macrophages that intact CD47 bestows upon tumor cells. Leiomyosarcoma (LMS) is a tumor of smooth muscle that can express varying levels of colony-stimulating factor-1 (CSF1), the expression of which correlates with the numbers of tumor-associated macrophages (TAMs) that are found in these tumors. We have previously shown that the presence of TAMs in LMS is associated with poor clinical outcome and the overall effect of TAMs in LMS therefore appears to be protumorigenic. However, the use of inhibitory antibodies against CD47 offers an opportunity to turn TAMs against LMS cells by allowing the phagocytic behavior of resident macrophages to predominate. Here we show that interference with CD47 increases phagocytosis of two human LMS cell lines, LMS04 and LMS05, in vitro. In addition, treatment of mice bearing subcutaneous LMS04 and LMS05 tumors with a novel, humanized anti-CD47 antibody resulted in significant reductions in tumor size. Mice bearing LMS04 tumors develop large numbers of lymph node and lung metastases. In a unique model for neoadjuvant treatment, mice were treated with anti-CD47 antibody starting 1 wk before resection of established primary tumors and subsequently showed a striking decrease in the size and number of metastases. These data suggest that treatment with anti-CD47 antibodies not only reduces primary tumor size but can also be used to inhibit the development of, or to eliminate, metastatic disease.
- Published
- 2012
- Full Text
- View/download PDF
44. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors.
- Author
-
Willingham SB, Volkmer JP, Gentles AJ, Sahoo D, Dalerba P, Mitra SS, Wang J, Contreras-Trujillo H, Martin R, Cohen JD, Lovelace P, Scheeren FA, Chao MP, Weiskopf K, Tang C, Volkmer AK, Naik TJ, Storm TA, Mosley AR, Edris B, Schmid SM, Sun CK, Chua MS, Murillo O, Rajendran P, Cha AC, Chin RK, Kim D, Adorno M, Raveh T, Tseng D, Jaiswal S, Enger PØ, Steinberg GK, Li G, So SK, Majeti R, Harsh GR, van de Rijn M, Teng NN, Sunwoo JB, Alizadeh AA, Clarke MF, and Weissman IL
- Subjects
- Antibodies immunology, CD47 Antigen genetics, Cell Division immunology, Flow Cytometry, Humans, Neoplasms pathology, Neoplasms therapy, Phagocytosis immunology, Prognosis, Survival Analysis, Antigens, Differentiation metabolism, CD47 Antigen immunology, Neoplasms immunology, RNA, Messenger genetics, Receptors, Immunologic metabolism
- Abstract
CD47, a "don't eat me" signal for phagocytic cells, is expressed on the surface of all human solid tumor cells. Analysis of patient tumor and matched adjacent normal (nontumor) tissue revealed that CD47 is overexpressed on cancer cells. CD47 mRNA expression levels correlated with a decreased probability of survival for multiple types of cancer. CD47 is a ligand for SIRPα, a protein expressed on macrophages and dendritic cells. In vitro, blockade of CD47 signaling using targeted monoclonal antibodies enabled macrophage phagocytosis of tumor cells that were otherwise protected. Administration of anti-CD47 antibodies inhibited tumor growth in orthotopic immunodeficient mouse xenotransplantation models established with patient tumor cells and increased the survival of the mice over time. Anti-CD47 antibody therapy initiated on larger tumors inhibited tumor growth and prevented or treated metastasis, but initiation of the therapy on smaller tumors was potentially curative. The safety and efficacy of targeting CD47 was further tested and validated in immune competent hosts using an orthotopic mouse breast cancer model. These results suggest all human solid tumor cells require CD47 expression to suppress phagocytic innate immune surveillance and elimination. These data, taken together with similar findings with other human neoplasms, show that CD47 is a commonly expressed molecule on all cancers, its function to block phagocytosis is known, and blockade of its function leads to tumor cell phagocytosis and elimination. CD47 is therefore a validated target for cancer therapies.
- Published
- 2012
- Full Text
- View/download PDF
45. Stimulation of natural killer cells with a CD137-specific antibody enhances trastuzumab efficacy in xenotransplant models of breast cancer.
- Author
-
Kohrt HE, Houot R, Weiskopf K, Goldstein MJ, Scheeren F, Czerwinski D, Colevas AD, Weng WK, Clarke MF, Carlson RW, Stockdale FE, Mollick JA, Chen L, and Levy R
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Breast Neoplasms drug therapy, Drug Synergism, Female, Humans, Mammary Neoplasms, Animal drug therapy, Mice, Mice, Nude, Mice, SCID, Neoplasm Transplantation, Transplantation, Heterologous, Trastuzumab, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal, Humanized administration & dosage, Killer Cells, Natural cytology, Tumor Necrosis Factor Receptor Superfamily, Member 9 metabolism
- Abstract
Trastuzumab, a monoclonal antibody targeting human epidermal growth factor receptor 2 (HER2; also known as HER-2/neu), is indicated for the treatment of women with either early stage or metastatic HER2(+) breast cancer. It kills tumor cells by several mechanisms, including antibody-dependent cellular cytotoxicity (ADCC). Strategies that enhance the activity of ADCC effectors, including NK cells, may improve the efficacy of trastuzumab. Here, we have shown that upon encountering trastuzumab-coated, HER2-overexpressing breast cancer cells, human NK cells become activated and express the costimulatory receptor CD137. CD137 activation, which was dependent on NK cell expression of the FcγRIII receptor, occurred both in vitro and in the peripheral blood of women with HER2-expressing breast cancer after trastuzumab treatment. Stimulation of trastuzumab-activated human NK cells with an agonistic mAb specific for CD137 killed breast cancer cells (including an intrinsically trastuzumab-resistant cell line) more efficiently both in vitro and in vivo in xenotransplant models of human breast cancer, including one using a human primary breast tumor. The enhanced cytotoxicity was restricted to antibody-coated tumor cells. This sequential antibody strategy, combining a tumor-targeting antibody with a second antibody that activates the host innate immune system, may improve the therapeutic effects of antibodies against breast cancer and other HER2-expressing tumors.
- Published
- 2012
- Full Text
- View/download PDF
46. CD137 stimulation enhances the antilymphoma activity of anti-CD20 antibodies.
- Author
-
Kohrt HE, Houot R, Goldstein MJ, Weiskopf K, Alizadeh AA, Brody J, Müller A, Pachynski R, Czerwinski D, Coutre S, Chao MP, Chen L, Tedder TF, and Levy R
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal, Murine-Derived pharmacology, Antibody-Dependent Cell Cytotoxicity, Drug Synergism, Humans, Rituximab, Tumor Cells, Cultured, Tumor Necrosis Factor Receptor Superfamily, Member 9 genetics, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Antigens, CD20 immunology, Tumor Necrosis Factor Receptor Superfamily, Member 9 immunology
- Abstract
Antibody-dependent cell-mediated cytotoxicity (ADCC), which is largely mediated by natural killer (NK) cells, is thought to play an important role in the efficacy of rituximab, an anti-CD20 monoclonal antibody (mAb) used to treat patients with B-cell lymphomas. CD137 is a costimulatory molecule expressed on a variety of immune cells after activation, including NK cells. In the present study, we show that an anti-CD137 agonistic mAb enhances the antilymphoma activity of rituximab by enhancing ADCC. Human NK cells up-regulate CD137 after encountering rituximab-coated tumor B cells, and subsequent stimulation of these NK cells with anti-CD137 mAb enhances rituximab-dependent cytotoxicity against the lymphoma cells. In a syngeneic murine lymphoma model and in a xenotransplanted human lymphoma model, sequential administration of anti-CD20 mAb followed by anti-CD137 mAb had potent antilymphoma activity in vivo. These results support a novel, sequential antibody approach against B-cell malignancies by targeting first the tumor and then the host immune system.
- Published
- 2011
- Full Text
- View/download PDF
47. Calreticulin is the dominant pro-phagocytic signal on multiple human cancers and is counterbalanced by CD47.
- Author
-
Chao MP, Jaiswal S, Weissman-Tsukamoto R, Alizadeh AA, Gentles AJ, Volkmer J, Weiskopf K, Willingham SB, Raveh T, Park CY, Majeti R, and Weissman IL
- Subjects
- Animals, Cell Line, Cell Line, Tumor, Flow Cytometry, Humans, In Vitro Techniques, Leukemia, Myeloid, Acute metabolism, Lymphoma, Non-Hodgkin metabolism, Mice, Urinary Bladder Neoplasms metabolism, CD47 Antigen metabolism, Calreticulin metabolism, Phagocytosis physiology
- Abstract
Under normal physiological conditions, cellular homeostasis is partly regulated by a balance of pro- and anti-phagocytic signals. CD47, which prevents cancer cell phagocytosis by the innate immune system, is highly expressed on several human cancers including acute myeloid leukemia, non-Hodgkin's lymphoma, and bladder cancer. Blocking CD47 with a monoclonal antibody results in phagocytosis of cancer cells and leads to in vivo tumor elimination, yet normal cells remain mostly unaffected. Thus, we postulated that cancer cells must also display a potent pro-phagocytic signal. Here, we identified calreticulin as a pro-phagocytic signal that was highly expressed on the surface of several human cancers, but was minimally expressed on most normal cells. Increased CD47 expression correlated with high amounts of calreticulin on cancer cells and was necessary for protection from calreticulin-mediated phagocytosis. Blocking the interaction of target cell calreticulin with its receptor, low-density lipoprotein receptor-related protein, on phagocytic cells prevented anti-CD47 antibody-mediated phagocytosis. Furthermore, increased calreticulin expression was an adverse prognostic factor in diverse tumors including neuroblastoma, bladder cancer, and non-Hodgkin's lymphoma. These findings identify calreticulin as the dominant pro-phagocytic signal on several human cancers, provide an explanation for the selective targeting of tumor cells by anti-CD47 antibody, and highlight the balance between pro- and anti-phagocytic signals in the immune evasion of cancer.
- Published
- 2010
- Full Text
- View/download PDF
48. Asterless is a scaffold for the onset of centriole assembly.
- Author
-
Dzhindzhev NS, Yu QD, Weiskopf K, Tzolovsky G, Cunha-Ferreira I, Riparbelli M, Rodrigues-Martins A, Bettencourt-Dias M, Callaini G, and Glover DM
- Subjects
- Animals, Animals, Genetically Modified, Cell Line, Centrosome metabolism, Drosophila Proteins chemistry, Drosophila Proteins deficiency, Drosophila Proteins genetics, Drosophila melanogaster cytology, Drosophila melanogaster embryology, Drosophila melanogaster genetics, Drosophila melanogaster metabolism, Female, Humans, Microtubule-Associated Proteins metabolism, Microtubule-Organizing Center metabolism, Oocytes cytology, Oocytes metabolism, Protein Binding, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases deficiency, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Cell Cycle Proteins metabolism, Centrioles metabolism, Drosophila Proteins metabolism
- Abstract
Centrioles are found in the centrosome core and, as basal bodies, at the base of cilia and flagella. Centriole assembly and duplication is controlled by Polo-like-kinase 4 (Plk4): these processes fail if Plk4 is downregulated and are promoted by Plk4 overexpression. Here we show that the centriolar protein Asterless (Asl; human orthologue CEP152) provides a conserved molecular platform, the amino terminus of which interacts with the cryptic Polo box of Plk4 whereas the carboxy terminus interacts with the centriolar protein Sas-4 (CPAP in humans). Drosophila Asl and human CEP152 are required for the centrosomal loading of Plk4 in Drosophila and CPAP in human cells, respectively. Depletion of Asl or CEP152 caused failure of centrosome duplication; their overexpression led to de novo centriole formation in Drosophila eggs, duplication of free centrosomes in Drosophila embryos, and centrosome amplification in cultured Drosophila and human cells. Overexpression of a Plk4-binding-deficient mutant of Asl prevented centriole duplication in cultured cells and embryos. However, this mutant protein was able to promote microtubule organizing centre (MTOC) formation in both embryos and oocytes. Such MTOCs had pericentriolar material and the centriolar protein Sas-4, but no centrioles at their core. Formation of such acentriolar MTOCs could be phenocopied by overexpression of Sas-4 in oocytes or embryos. Our findings identify independent functions for Asl as a scaffold for Plk4 and Sas-4 that facilitates self-assembly and duplication of the centriole and organization of pericentriolar material.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.