1
|
Niu L, Jang E, Chin AL, Huo Z, Wang W, Cai W, Tong R. Noncovalently particle-anchored cytokines with prolonged tumor retention safely elicit potent antitumor immunity. SCIENCE ADVANCES 2024; 10:eadk7695. [PMID: 38640236 PMCID: PMC11029804 DOI: 10.1126/sciadv.adk7695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 03/19/2024] [Indexed: 04/21/2024]
Abstract
Preclinical studies have shown that immunostimulatory cytokines elicit antitumor immune responses but their clinical use is limited by severe immune-related adverse events upon systemic administration. Here, we report a facile and versatile strategy for noncovalently anchoring potent Fc-fused cytokine molecules to the surface of size-discrete particles decorated with Fc-binding peptide for local administration. Following intratumoral injection, particle-anchored Fc cytokines exhibit size-dependent intratumoral retention. The 1-micrometer particle prolongs intratumoral retention of Fc cytokine for over a week and has minimal systemic exposure, thereby eliciting antitumor immunity while eliminating systemic toxicity caused by circulating cytokines. In addition, the combination of these particle-anchored cytokines with immune checkpoint blockade antibodies safely promotes tumor regression in various syngeneic tumor models and genetically engineered murine tumor models and elicits systemic antitumor immunity against tumor rechallenge. Our formulation strategy renders a safe and tumor-agnostic approach that uncouples cytokines' immunostimulatory properties from their systemic toxicities for potential clinical application.
Collapse
Affiliation(s)
- Liqian Niu
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Eungyo Jang
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Ai Lin Chin
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Ziyu Huo
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| | - Wenbo Wang
- Department of Materials Science and Engineering, Virginia Polytechnic Institute and State University, 445 Old Turner Street, Blacksburg, VA, 24061, USA
| | - Wenjun Cai
- Department of Materials Science and Engineering, Virginia Polytechnic Institute and State University, 445 Old Turner Street, Blacksburg, VA, 24061, USA
| | - Rong Tong
- Department of Chemical Engineering, Virginia Polytechnic Institute and State University, 635 Prices Fork Road, Blacksburg, VA, 24061, USA
| |
Collapse
|
2
|
Andrechak JC, Dooling LJ, Tobin MP, Zhang W, Hayes BH, Lee JY, Jin X, Irianto J, Discher DE. CD47-SIRPα Checkpoint Disruption in Metastases Requires Tumor-Targeting Antibody for Molecular and Engineered Macrophage Therapies. Cancers (Basel) 2022; 14:1930. [PMID: 35454837 PMCID: PMC9026896 DOI: 10.3390/cancers14081930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 11/17/2022] Open
Abstract
The macrophage checkpoint interaction CD47-SIRPα is an emerging target for cancer therapy, but clinical trials of monoclonal anti-CD47 show efficacy only in liquid tumors when combined with tumor-opsonizing IgG. Here, in challenging metastatic solid tumors, CD47 deletion shows no effect on tumor growth unless combined with otherwise ineffective tumor-opsonization, and we likewise show wild-type metastases are suppressed by SIRPα-blocked macrophages plus tumor-opsonization. Lung tumor nodules of syngeneic B16F10 melanoma cells with CD47 deletion show opsonization drives macrophage phagocytosis of B16F10s, consistent with growth versus phagocytosis calculus for exponential suppression of cancer. Wild-type CD47 levels on metastases in lungs of immunocompetent mice and on human metastases in livers of immunodeficient mice show that systemic injection of antibody-engineered macrophages also suppresses growth. Such in vivo functionality can be modulated by particle pre-loading of the macrophages. Thus, even though CD47-SIRPα disruption and tumor-opsonizing IgG are separately ineffective against established metastatic solid tumors, their combination in molecular and cellular therapies prolongs survival.
Collapse
Affiliation(s)
- Jason C. Andrechak
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lawrence J. Dooling
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
| | - Michael P. Tobin
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William Zhang
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brandon H. Hayes
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justine Y. Lee
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
| | - Xiaoling Jin
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
| | - Jerome Irianto
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Dennis E. Discher
- Biophysical Engineering Labs, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.C.A.); (L.J.D.); (M.P.T.); (W.Z.); (B.H.H.); (J.Y.L.); (X.J.); (J.I.)
- Graduate Group of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Yura Y, Hamada M. Oral Immune-Related Adverse Events Caused by Immune Checkpoint Inhibitors: Salivary Gland Dysfunction and Mucosal Diseases. Cancers (Basel) 2022; 14:cancers14030792. [PMID: 35159059 PMCID: PMC8834130 DOI: 10.3390/cancers14030792] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 01/29/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Conventional chemotherapy and targeted therapies have limited efficacy against advanced head and neck squamous cell carcinoma (HNSCC). The immune checkpoint inhibitors (ICIs) such as antibodies against CTLA-4, PD-1, and PD-L1 interrupt the co-inhibitory pathway of T cells and enhance the ability of CD8+ T cells to destroy tumors. Even in advanced HNSCC patients with recurrent diseases and distant metastasis, ICI therapy shows efficiency and become an effective alternative to conventional chemotherapy. However, as this therapy releases the immune tolerance state, cytotoxic CD8+ T cells can also attack organs and tissues expressing self-antigens that cross-react with tumor antigens and induce immune-related adverse events (irAEs). When patients with HNSCC are treated with ICIs, autoimmune diseases occur in multiple organs including the skin, digestive tract, endocrine system, liver, and respiratory tract. Treatment of various malignancies, including HNSCC, with ICIs may result in the appearance of oral irAEs. In the oral cavity, an oral lichenoid reaction (OLR) and pemphigoid develop. Sicca syndrome also occurs in association with ICIs, affecting the salivary glands to induce xerostomia. It is necessary to elucidate the pathogenic mechanisms of these intractable diseases that are not seen with conventional therapy. Early diagnosis and appropriate approaches to irAEs are needed for efficient treatment of advanced HNSCC by ICIs.
Collapse
|
4
|
Gautron A, Migault M, Bachelot L, Corre S, Galibert MD, Gilot D. Human TYRP1: Two functions for a single gene? Pigment Cell Melanoma Res 2021; 34:836-852. [PMID: 33305505 DOI: 10.1111/pcmr.12951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 11/12/2020] [Accepted: 12/01/2020] [Indexed: 01/07/2023]
Abstract
In the animal kingdom, skin pigmentation is highly variable between species, and it contributes to phenotypes. In humans, skin pigmentation plays a part in sun protection. Skin pigmentation depends on the ratio of the two pigments pheomelanin and eumelanin, both synthesized by a specialized cell population, the melanocytes. In this review, we explore one important factor in pigmentation: the tyrosinase-related protein 1 (TYRP1) gene which is involved in eumelanin synthesis via the TYRP1 protein. Counterintuitively, high TYRP1 mRNA expression is associated with a poor clinical outcome for patients with metastatic melanomas. Recently, we were able to explain this unexpected TYRP1 function by demonstrating that TYRP1 mRNA sequesters microRNA-16, a tumor suppressor miRNA. Here, we focus on actors influencing TYRP1 mRNA abundance, particularly transcription factors, single nucleotide polymorphisms (SNPs), and miRNAs, as they all dictate the indirect oncogenic activity of TYRP1.
Collapse
Affiliation(s)
- Arthur Gautron
- CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Univ. Rennes, Rennes, France
| | - Mélodie Migault
- CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Univ. Rennes, Rennes, France.,Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Laura Bachelot
- CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Univ. Rennes, Rennes, France
| | - Sébastien Corre
- CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Univ. Rennes, Rennes, France
| | - Marie-Dominique Galibert
- CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Univ. Rennes, Rennes, France.,CHU Rennes, Génétique Moléculaire et Génomique, UMR 6290, F-35000, Rennes, France
| | - David Gilot
- CNRS, IGDR (Institut de génétique et développement de Rennes) - UMR 6290, F-35000, Univ. Rennes, Rennes, France.,INSERM U1242, Centre Eugène Marquis, Rennes, France
| |
Collapse
|
5
|
Broadway KM, Scharf BE. Salmonella Typhimurium as an Anticancer Therapy: Recent Advances and Perspectives. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019. [DOI: 10.1007/s40588-019-00132-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
6
|
Melanoma and Vitiligo: In Good Company. Int J Mol Sci 2019; 20:ijms20225731. [PMID: 31731645 PMCID: PMC6888090 DOI: 10.3390/ijms20225731] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/10/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022] Open
Abstract
Cutaneous melanoma represents the most aggressive form of skin cancer, whereas vitiligo is an autoimmune disorder that leads to progressive destruction of skin melanocytes. However, vitiligo has been associated with cutaneous melanoma since the 1970s. Most of the antigens recognized by the immune system are expressed by both melanoma cells and normal melanocytes, explaining why the autoimmune response against melanocytes that led to vitiligo could be also present in melanoma patients. Leukoderma has been also observed as a side effect of melanoma immunotherapy and has always been associated with a favorable prognosis. In this review, we discuss several characteristics of the immune system responses shared by melanoma and vitiligo patients, as well as the significance of occurrence of leukoderma during immunotherapy, with special attention to check-point inhibitors.
Collapse
|
7
|
Nigar S, Shimosato T. Cooperation of Oligodeoxynucleotides and Synthetic Molecules as Enhanced Immune Modulators. Front Nutr 2019; 6:140. [PMID: 31508424 PMCID: PMC6718720 DOI: 10.3389/fnut.2019.00140] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022] Open
Abstract
Unmethylated cytosine–guanine dinucleotide (CpG) motifs are potent stimulators of the host immune response. Cellular recognition of CpG motifs occurs via Toll-like receptor 9 (TLR9), which normally activates immune responses to pathogen-associated molecular patterns (PAMPs) indicative of infection. Oligodeoxynucleotides (ODNs) containing unmethylated CpGs mimic the immunostimulatory activity of viral/microbial DNA. Synthetic ODNs harboring CpG motifs resembling those identified in viral/microbial DNA trigger an identical response, such that these immunomodulatory ODNs have therapeutic potential. CpG DNA has been investigated as an agent for the management of malignancy, asthma, allergy, and contagious diseases, and as an adjuvant in immunotherapy. In this review, we discuss the potential synergy between synthetic ODNs and other synthetic molecules and their immunomodulatory effects. We also summarize the different synthetic molecules that function as immune modulators and outline the phenomenon of TLR-mediated immune responses. We previously reported a novel synthetic ODN that acts synergistically with other synthetic molecules (including CpG ODNs, the synthetic triacylated lipopeptide Pam3CSK4, lipopolysaccharide, and zymosan) that could serve as an immune therapy. Additionally, several clinical trials have evaluated the use of CpG ODNs with other immune factors such as granulocyte-macrophage colony-stimulating factor, cytokines, and both endosomal and cell-surface TLR ligands as adjuvants for the augmentation of vaccine activity. Furthermore, we discuss the structural recognition of ODNs by TLRs and the mechanism of functional modulation of TLRs in the context of the potential application of ODNs as wide-spectrum therapeutic agents.
Collapse
Affiliation(s)
- Shireen Nigar
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Takeshi Shimosato
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
8
|
Vaish U, Kumar AA, Varshney S, Ghosh S, Sengupta S, Sood C, Kar HK, Sharma P, Natarajan VT, Gokhale RS, Rani R. Micro RNAs upregulated in Vitiligo skin play an important role in its aetiopathogenesis by altering TRP1 expression and keratinocyte-melanocytes cross-talk. Sci Rep 2019; 9:10079. [PMID: 31300697 PMCID: PMC6625998 DOI: 10.1038/s41598-019-46529-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 07/01/2019] [Indexed: 01/20/2023] Open
Abstract
Translation of genes is regulated by many factors including microRNAs (miRNAs). miRNA profiling of lesional and non-lesional epidermal RNA from 18 vitiligo patients revealed significant upregulation of 29 miRNAs in the lesional epidermis, of which 6 miRNAs were transfected in normal human epidermal keratinocytes (NHEKs) to study their downstream effects using quantitative proteomics. Many proteins involved in oxidative stress, Vesicle trafficking, Cellular apoptosis, Mitochondrial proteins and Keratins were regulated after miRNA transfections in the keratinocytes. However, tyrosinase related protein-1 (TRP1/TYRP1), a melanogenesis protein, was consistently downregulated in NHEKs by all the six miRNAs tested, which was quite intriguing. TRP1 was also downregulated in lesional epidermis compared with non-lesional epidermis. Since melanocytes synthesize and transfer melanosomes to the surrounding keratinocytes, we hypothesized that downregulation of TRP1 in NHEKs may have a role in melanosome transfer, which was confirmed by our co-culture experiments. Downregulation of TRP1 in keratinocytes negatively affected the melanosome transfer from melanocytes to keratinocytes resulting in melanin accumulation which may be leading to melanin induced cytotoxicity in melanocytes. Regulation of key processes involved in aetiopathogenesis of vitiligo along with TRP1 suggests that miRNAs act in an integrated manner which may be detrimental for the loss of melanocytes in vitiligo.
Collapse
Affiliation(s)
| | | | - Swati Varshney
- CSIR-Institute of Genomics & Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-IGIB, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India
| | - Shreya Ghosh
- CSIR-Institute of Genomics & Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India
| | - Shantanu Sengupta
- CSIR-Institute of Genomics & Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-IGIB, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India
| | - Chandni Sood
- National Institute of Immunology, New Delhi, 110067, India
| | - Hemanta K Kar
- Dr. Ram Manohar Lohia Hospital, New Delhi, 110001, India
| | - Pankaj Sharma
- Dr. Ram Manohar Lohia Hospital, New Delhi, 110001, India
| | - Vivek T Natarajan
- CSIR-Institute of Genomics & Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-IGIB, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India
| | - Rajesh S Gokhale
- National Institute of Immunology, New Delhi, 110067, India.,CSIR-Institute of Genomics & Integrative Biology, Mathura Road, Sukhdev Vihar, New Delhi, 110025, India
| | - Rajni Rani
- National Institute of Immunology, New Delhi, 110067, India.
| |
Collapse
|
9
|
Fässler M, Diem S, Mangana J, Hasan Ali O, Berner F, Bomze D, Ring S, Niederer R, Del Carmen Gil Cruz C, Pérez Shibayama CI, Krolik M, Siano M, Joerger M, Recher M, Risch L, Güsewell S, Risch M, Speiser DE, Ludewig B, Levesque MP, Dummer R, Flatz L. Antibodies as biomarker candidates for response and survival to checkpoint inhibitors in melanoma patients. J Immunother Cancer 2019; 7:50. [PMID: 30786924 PMCID: PMC6383238 DOI: 10.1186/s40425-019-0523-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/31/2019] [Indexed: 01/09/2023] Open
Abstract
Background Long-term survival of stage IV melanoma patients has improved significantly with the development of immune checkpoint inhibitors (CIs). Reliable biomarkers to predict response and clinical outcome are needed. Methods We investigated the role of melanoma-associated antibodies as predictive markers for CI therapy in two independent cohorts. In cohort 1, a prospective study, we measured specific antibodies before treatment, after one week and after six to nine weeks of treatment. Cohort 2 consisted of serum samples prior to CI therapy initiation. ELISA assays were performed to quantify specific IgG directed against melanocyte differentiation antigens tyrosinase-related proteins 1 and 2 (TRP1/TYRP1 and TRP2/TYRP2), glycoprotein 100 (gp100), MelanA/MART1, and the cancer-testis antigen NY-ESO-1. Response was defined as either complete or partial remission on CT scan according to RECIST 1.1. Results In cohort 1, baseline levels of these antibodies were higher in the responder group, although statistical significance was only reached for NY-ESO-1 (p = 0.007). In cohort 2, significantly higher antibody baseline levels for MelanA/MART1 (p = 0.003) and gp100 (p = 0.029) were found. After pooling the results from both cohorts, higher levels of MelanA/MART1 (p = 0.013), TRP1/TYRP1 (p = 0.048), TRP2/TYRP2 (p = 0.047) and NY-ESO-1 (p = 0.005) specific antibodies at baseline were independently associated with response. Conclusions Melanoma-associated antibodies may be candidate biomarkers for response and survival in metastatic melanoma patients being treated with CIs. These markers may be used to complement patient assessment, in combination with PD-L1 status, tumor-infiltrating lymphocytes and tumor mutational burden, with the aim to predict outcome of CI treatment in patients with metastatic melanoma. Trial registration Ethikkommission Ostschweiz, EKOS 16/079 https://ongoingprojects.swissethics.ch/runningProjects_list.php?q=%28BASECID~contains~2016-00998%29&orderby=dBASECID. Electronic supplementary material The online version of this article (10.1186/s40425-019-0523-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mirjam Fässler
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland.,Department of Dermatology, Allergology and Venerology, Kantonsspital St.Gallen, Rorschacher Str. 95, 9007, St. Gallen, Switzerland
| | - Stefan Diem
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland.,Department of Oncology/Hematology, Kantonsspital St.Gallen, Rorschacher Str. 95, 9007, St. Gallen, Switzerland.,Department of Oncology/Hematology, Spital Grabs, Spitalstrasse 44, 9472, Grabs, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Omar Hasan Ali
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland.,Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Fiamma Berner
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
| | - David Bomze
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
| | - Sandra Ring
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
| | - Rebekka Niederer
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
| | | | | | - Michal Krolik
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
| | - Marco Siano
- Department of Oncology/Hematology, Kantonsspital St.Gallen, Rorschacher Str. 95, 9007, St. Gallen, Switzerland
| | - Markus Joerger
- Department of Oncology/Hematology, Kantonsspital St.Gallen, Rorschacher Str. 95, 9007, St. Gallen, Switzerland
| | - Mike Recher
- Clinic for Primary Immunodeficiency, Medical Outpatient Unit and Immunodeficiency Laboratory, Department of Biomedicine, University Hospital, Hebelstrasse 20, 4067, Basel, Switzerland
| | - Lorenz Risch
- Labormedizinisches Zentrum Dr. Risch Ostschweiz AG, Brauerstrasse 95, 9016, St. Gallen, Switzerland.,Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, INO-F, 3010, Bern, Switzerland.,Private University Triesen, Dorfstrasse 24, 9495, Triesen, Liechtenstein
| | - Sabine Güsewell
- Clinical Trials Unit, Kantonsspital St.Gallen, Bedastrasse 1, 9000, St. Gallen, Switzerland
| | - Martin Risch
- Labormedizinisches Zentrum Dr. Risch Ostschweiz AG, Brauerstrasse 95, 9016, St. Gallen, Switzerland.,Department of Laboratory Medicine, Kantonsspital Graubünden, Loestrasse 170, 7000, Chur, Switzerland
| | - Daniel E Speiser
- Ludwig Cancer Research, University of Lausanne, Chemin des Boveresses 155, 1066 Épalinges, Lausanne, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Lukas Flatz
- Institute of Immunobiology, Kantonsspital St.Gallen, Rorschacherstrasse 95, 9007, St. Gallen, Switzerland. .,Department of Dermatology, Allergology and Venerology, Kantonsspital St.Gallen, Rorschacher Str. 95, 9007, St. Gallen, Switzerland. .,Department of Oncology/Hematology, Kantonsspital St.Gallen, Rorschacher Str. 95, 9007, St. Gallen, Switzerland. .,Department of Dermatology, University Hospital Zurich, University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland. .,Clinical Trials Unit, Kantonsspital St.Gallen, Bedastrasse 1, 9000, St. Gallen, Switzerland.
| |
Collapse
|
10
|
Breaking tolerance with engineered class I antigen-presenting molecules. Proc Natl Acad Sci U S A 2019; 116:3136-3145. [PMID: 30728302 DOI: 10.1073/pnas.1807465116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Successful efforts to activate T cells capable of recognizing weak cancer-associated self-antigens have employed altered peptide antigens to activate T cell responses capable of cross-reacting on native tumor-associated self. A limitation of this approach is the requirement for detailed knowledge about the altered self-peptide ligands used in these vaccines. In the current study we considered allorecognition as an approach for activating CTL capable of recognizing weak or self-antigens in the context of self-MHC. Nonself antigen-presenting molecules typically contain polymorphisms that influence interactions with the bound peptide and TCR interface. Recognition of these nonself structures results in peptide-dependent alloimmunity. Alloreactive T cells target their inducing alloantigens as well as third-party alloantigens but generally fail to target self-antigens. Certain residues located on the alpha-1/2 domains of class I antigen-presenting molecules primarily interface with TCR. These residues are more conserved within and across species than are residues that determine peptide antigen binding properties. Class I variants designed with amino acid substitutions at key positions within the conserved helical structures are shown to provide strong activating signals to alloreactive CD8 T cells while avoiding changes in naturally bound peptide ligands. Importantly, CTL activated in this manner can break self-tolerance by reacting to self-peptides presented by native MHC. The ability to activate self-tolerant T cells capable of cross-reacting on self-peptide-MHC in vivo represents an approach for inducing autoimmunity, with possible application in cancer vaccines.
Collapse
|
11
|
Benonisson H, Sow HS, Breukel C, Claassens J, Brouwers C, Linssen MM, Fransen MF, Sluijter M, Ossendorp F, van Hall T, Verbeek JS. High FcγR Expression on Intratumoral Macrophages Enhances Tumor-Targeting Antibody Therapy. THE JOURNAL OF IMMUNOLOGY 2018; 201:3741-3749. [PMID: 30397036 DOI: 10.4049/jimmunol.1800700] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/05/2018] [Indexed: 12/17/2022]
Abstract
Therapy with tumor-specific Abs is common in the clinic but has limited success against solid malignancies. We aimed at improving the efficacy of this therapy by combining a tumor-specific Ab with immune-activating compounds. In this study, we demonstrate in the aggressive B16F10 mouse melanoma model that concomitant application of the anti-TRP1 Ab (clone TA99) with TLR3-7/8 or -9 ligands, and IL-2 strongly enhanced tumor control in a therapeutic setting. Depletion of NK cells, macrophages, or CD8+ T cells all mitigated the therapeutic response, showing a coordinated immune rejection by innate and adaptive immune cells. FcγRs were essential for the therapeutic effect, with a dominant role for FcγRI and a minor role for FcγRIII and FcγRIV. FcγR expression on NK cells and granulocytes was dispensable, indicating that other tumoricidal functions of NK cells were involved and implicating that FcγRI, -III, and -IV exerted their activity on macrophages. Indeed, F4/80+Ly-6C+ inflammatory macrophages in the tumor microenvironment displayed high levels of these receptors. Whereas administration of the anti-TRP1 Ab alone reduced the frequency of these macrophages, the combination with a TLR agonist retained these cells in the tumor microenvironment. Thus, the addition of innate stimulatory compounds, such as TLR ligands, to tumor-specific Ab therapy could greatly enhance its efficacy in solid cancers via optimal exploitation of FcγRs.
Collapse
Affiliation(s)
- Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Heng Sheng Sow
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Cor Breukel
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Jill Claassens
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Conny Brouwers
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Margot M Linssen
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands; and
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands;
| |
Collapse
|
12
|
Benonisson H, Altıntaş I, Sluijter M, Verploegen S, Labrijn AF, Schuurhuis DH, Houtkamp MA, Verbeek JS, Schuurman J, van Hall T. CD3-Bispecific Antibody Therapy Turns Solid Tumors into Inflammatory Sites but Does Not Install Protective Memory. Mol Cancer Ther 2018; 18:312-322. [PMID: 30381448 DOI: 10.1158/1535-7163.mct-18-0679] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/01/2018] [Accepted: 10/24/2018] [Indexed: 11/16/2022]
Abstract
Immunotherapy of cancer with CD3-targeting bispecific antibodies (CD3 bsAb) is a fast developing field, and multiple tumor-associated antigens (TAA) are evaluated for hematologic and solid malignancies. The efficacy of these CD3 bsAb is usually examined in xenograft mouse tumor models with human T cells or in genetically engineered mouse models, where human TAA are introduced. These models often fail to fully recapitulate the natural tumor environment, especially for solid cancers, because of interspecies differences. Here, we investigated the systemic and intratumoral effects of a mouse CD3 bsAb in a fully immune-competent mouse melanoma model. Systemic administration of 0.5 mg/kg antibody induced a brief overall T-cell activation that was selectively sustained in the tumor microenvironment for several days. A fast subsequent influx of inflammatory macrophages into the tumor microenvironment was observed, followed by an increase in the number of CD4+ and CD8+ T cells. Although the capacity to directly kill melanoma cells in vitro was very modest, optimal tumor elimination was observed in vivo, even in the absence of CD8+ T cells, implying a redundancy in T-cell subsets for therapeutic efficacy. Finally, we took advantage of the full immune competence of our mouse model and tested immune memory induction. Despite a strong initial immunity against melanoma, treatment with the CD3 bsAb did not install protective memory responses. The observed mechanisms of action revealed in this immune-competent mouse model might form a rational basis for combinatorial approaches.
Collapse
Affiliation(s)
- Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
13
|
Kumar S, Singh R, Malik S, Manne U, Mishra M. Prostate cancer health disparities: An immuno-biological perspective. Cancer Lett 2018; 414:153-165. [PMID: 29154974 PMCID: PMC5743619 DOI: 10.1016/j.canlet.2017.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/09/2017] [Accepted: 11/11/2017] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa) is the most commonly diagnosed malignancy in males, and, in the United States, is the second leading cause of cancer-related death for men older than 40 years. There is a higher incidence of PCa for African Americans (AAs) than for European-Americans (EAs). Investigations related to the incidence of PCa-related health disparities for AAs suggest that there are differences in the genetic makeup of these populations. Other differences are environmentally induced (e.g., diet and lifestyle), and the exposures are different. Men who immigrate from Eastern to Western countries have a higher risk of PCa than men in their native countries. However, the number of immigrants developing PCa is still lower than that of men in Western countries, suggesting that genetic factors are involved in the development of PCa. Altered genetic polymorphisms are associated with PCa progression. Androgens and the androgen receptor (AR) are involved in the development and progression of PCa. For populations with diverse racial/ethnic backgrounds, differences in lifestyle, diet, and biology, including genetic mutations/polymorphisms and levels of androgens and AR, are risk factors for PCa. Here, we provide an immuno-biological perspective on PCa in relation to racial/ethnic disparities and identify factors associated with the disproportionate incidence of PCa and its clinical outcomes.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Shalie Malik
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA; Department of Zoology, University of Lucknow, Lucknow 226007, India
| | - Upender Manne
- Department of Pathology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Manoj Mishra
- Cancer Biology Research and Training Program, Department of Biological Sciences, Alabama State University, Montgomery, AL 36104, USA.
| |
Collapse
|
14
|
Tam SH, McCarthy SG, Armstrong AA, Somani S, Wu SJ, Liu X, Gervais A, Ernst R, Saro D, Decker R, Luo J, Gilliland GL, Chiu ML, Scallon BJ. Functional, Biophysical, and Structural Characterization of Human IgG1 and IgG4 Fc Variants with Ablated Immune Functionality. Antibodies (Basel) 2017; 6:E12. [PMID: 31548527 PMCID: PMC6698826 DOI: 10.3390/antib6030012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 02/07/2023] Open
Abstract
Engineering of fragment crystallizable (Fc) domains of therapeutic immunoglobulin (IgG) antibodies to eliminate their immune effector functions while retaining other Fc characteristics has numerous applications, including blocking antigens on Fc gamma (Fcγ) receptor-expressing immune cells. We previously reported on a human IgG2 variant termed IgG2σ with barely detectable activity in antibody-dependent cellular cytotoxicity, phagocytosis, complement activity, and Fcγ receptor binding assays. Here, we extend that work to IgG1 and IgG4 antibodies, alternative subtypes which may offer advantages over IgG2 antibodies. In several in vitro and in vivo assays, the IgG1σ and IgG4σ variants showed equal or even lower Fc-related activities than the corresponding IgG2σ variant. In particular, IgG1σ and IgG4σ variants demonstrate complete lack of effector function as measured by antibody-dependent cellular cytotoxicity, complement-dependent cytotoxicity, antibody-dependent cellular phagocytosis, and in vivo T-cell activation. The IgG1σ and IgG4σ variants showed acceptable solubility and stability, and typical human IgG1 pharmacokinetic profiles in human FcRn-transgenic mice and cynomolgus monkeys. In silico T-cell epitope analyses predict a lack of immunogenicity in humans. Finally, crystal structures and simulations of the IgG1σ and IgG4σ Fc domains can explain the lack of Fc-mediated immune functions. These variants show promise for use in those therapeutic antibodies and Fc fusions for which the Fc domain should be immunologically "silent".
Collapse
Affiliation(s)
- Susan H Tam
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Stephen G McCarthy
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Anthony A Armstrong
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Sandeep Somani
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Sheng-Jiun Wu
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Xuesong Liu
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Alexis Gervais
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Robin Ernst
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Dorina Saro
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Rose Decker
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Jinquan Luo
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Gary L Gilliland
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Mark L Chiu
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| | - Bernard J Scallon
- Janssen Research & Development, LLC, 1400 McKean Road, Spring House, Ambler, PA 19477, USA.
| |
Collapse
|
15
|
Braster R, Grewal S, Visser R, Einarsdottir HK, van Egmond M, Vidarsson G, Bögels M. Human IgG3 with extended half-life does not improve Fc-gamma receptor-mediated cancer antibody therapies in mice. PLoS One 2017; 12:e0177736. [PMID: 28542406 PMCID: PMC5438146 DOI: 10.1371/journal.pone.0177736] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 05/02/2017] [Indexed: 01/01/2023] Open
Abstract
Background Current anti-cancer therapeutic antibodies that are used in the clinic are predominantly humanized or fully human immunoglobulin G1 (IgG1). These antibodies bind with high affinity to the target antigen and are efficient in activating the immune system via IgG Fc receptors and/or complement. In addition to IgG1, three more isotypes are present in humans, of which IgG3 has been found to be superior compared to human IgG1 in inducing antibody dependent cell cytotoxicity (ADCC), phagocytosis or activation of complement in some models. Nonetheless, no therapeutic human IgG3 mAbs have been developed due to the short in vivo half-life of most known IgG3 allotypes. In this manuscript, we compared the efficacy of V-gene matched IgG1 and IgG3 anti-tumour mAb (TA99) in mice, using natural variants of human IgG3 with short- or long half-life, differing only at position 435 with an arginine or histidine, respectively. Results In vitro human IgG1 and IgG3 did not show any differences in opsonisation ability of B16F10-gp75 mouse melanoma cells. IgG1, however, was superior in inducing phagocytosis of tumour cells by mouse macrophages. Similarly, in a mouse peritoneal metastasis model we did not detect an improved effect of IgG3 in preventing tumour outgrowth. Moreover, replacing the arginine at position 435 for a histidine in IgG3 to enhance half-life did not result in better suppression of tumour outgrowth compared to wild type IgG3 when injected prior to tumour cell injection. Conclusion In conclusion, human IgG3 does not have improved therapeutic efficacy compared to human IgG1 in a mouse tumour model.
Collapse
Affiliation(s)
- Rens Braster
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
| | - Simran Grewal
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Surgery, VU University Medical Centre, Amsterdam, The Netherlands
| | - Remco Visser
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Helga K. Einarsdottir
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Surgery, VU University Medical Centre, Amsterdam, The Netherlands
| | - Gestur Vidarsson
- Department of Experimental Immunohematology, Sanquin Research, and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
- * E-mail:
| | - Marijn Bögels
- Department of Molecular Cell Biology and Immunology, VU University Medical Centre, Amsterdam, The Netherlands
- Department of Surgery, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Khalil DN, Postow MA, Ibrahim N, Ludwig DL, Cosaert J, Kambhampati SRP, Tang S, Grebennik D, Kauh JSW, Lenz HJ, Flaherty KT, Hodi FS, Lawrence DP, Wolchok JD. An Open-Label, Dose-Escalation Phase I Study of Anti-TYRP1 Monoclonal Antibody IMC-20D7S for Patients with Relapsed or Refractory Melanoma. Clin Cancer Res 2016; 22:5204-5210. [PMID: 27797971 PMCID: PMC5117650 DOI: 10.1158/1078-0432.ccr-16-1241] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/04/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Tyrosinase-related protein-1 (TYRP1) is a transmembrane glycoprotein that is specifically expressed in melanocytes and melanoma cells. Preclinical data suggest that mAbs targeting TYRP1 confer antimelanoma activity. IMC-20D7S is a recombinant human IgG1 mAb targeting TYRP1. Here, we report the first-in-human phase I/Ib trial of IMC-20D7S. EXPERIMENTAL DESIGN The primary objective of this study was to establish the safety profile and the MTD of IMC-20D7S. Patients with advanced melanoma who progressed after or during at least one line of treatment or for whom standard therapy was not indicated enrolled in this standard 3 + 3 dose-escalation, open-label study. IMC-20D7S was administered intravenously every 2 or 3 weeks. RESULTS Twenty-seven patients were enrolled. The most common adverse events were fatigue and constipation experienced by nine (33%) and eight (30%) patients, respectively. There were no serious adverse events related to treatment, no discontinuations of treatment due to adverse events, and no treatment-related deaths. Given the absence of dose-limiting toxicities, an MTD was not defined, but a provisional MTD was established at the 20 mg/kg every 2-week dose based on serum concentration and safety data. One patient experienced a complete response. A disease control rate, defined as stable disease or better, of 41% was observed. CONCLUSION IMC-20D7S is well tolerated among patients with advanced melanoma with evidence of antitumor activity. Further investigation of this agent as monotherapy in selected patients or as part of combination regimens is warranted. Clin Cancer Res; 22(21); 5204-10. ©2016 AACR.
Collapse
Affiliation(s)
- Danny N Khalil
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York
| | | | | | | | | | | | | | | | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, Ludwig Center for Cancer Immunotherapy, New York, New York.
| |
Collapse
|
17
|
Guy TV, Terry AM, Bolton HA, Hancock DG, Shklovskaya E, Fazekas de St. Groth B. Pro- and anti-tumour effects of B cells and antibodies in cancer: a comparison of clinical studies and preclinical models. Cancer Immunol Immunother 2016; 65:885-96. [PMID: 27222052 PMCID: PMC11029718 DOI: 10.1007/s00262-016-1848-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 05/12/2016] [Indexed: 12/15/2022]
Abstract
The primary immune role of B cells is to produce antibodies, but they can also influence T cell function via antigen presentation and, in some contexts, immune regulation. Whether their roles in tumour immunity are similar to those in other chronic immune responses such as autoimmunity and chronic infection, where both pro- and anti-inflammatory roles have been described, remains controversial. Many studies have aimed to define the role of B cells in antitumor immune responses, but despite this considerable body of work, it is not yet possible to predict how they will affect immunity to any given tumour. In many human cancers, the presence of tumour-infiltrating B cells and tumour-reactive antibodies correlates with extended patient survival, and this clinical observation is supported by data from some animal models. On the other hand, T cell responses can be adversely affected by B cell production of immunoregulatory cytokines, a phenomenon that has been demonstrated in humans and in animal models. The isotype and concentration of tumour-reactive antibodies may also influence tumour progression. Recruitment of B cells into tumours may directly reflect the subtype and strength of the anti-tumour T cell response. As the response becomes chronic, B cells may attenuate T cell responses in an attempt to decrease host damage, similar to their described role in chronic infection and autoimmunity. Understanding how B cell responses in cancer are related to the effectiveness of the overall anti-tumour response is likely to aid in the development of new therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Thomas V Guy
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alexandra M Terry
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Holly A Bolton
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - David G Hancock
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Elena Shklovskaya
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Barbara Fazekas de St. Groth
- T Cell Biology Research Program, Centenary Institute of Cancer Medicine and Cell Biology, Locked Bag No. 6, Newtown, NSW, 2042, Australia.
- Discipline of Dermatology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
18
|
Chu D, Zhao Q, Yu J, Zhang F, Zhang H, Wang Z. Nanoparticle Targeting of Neutrophils for Improved Cancer Immunotherapy. Adv Healthc Mater 2016; 5:1088-93. [PMID: 26989887 DOI: 10.1002/adhm.201500998] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/07/2016] [Indexed: 11/08/2022]
Abstract
Cancer immunotherapy using tumor-specific monoclonal antibodies presents a novel approach for cancer treatment. A monoclonal antibody TA99 specific for gp75 antigen of melanoma initiates neutrophil recruitment in tumor responsible for cancer therapy. Here, a strategy is reported for hijacking neutrophils in vivo using nanoparticles (NPs) to deliver therapeutics into tumor. In a mouse model of melanoma, it is shown that systemically delivered albumin NPs increase in tumor when TA99 antibody is injected; and the NP tumor accumulation is mediated by neutrophils. After the administration of pyropheophorbide-a loaded albumin NPs and TA99, photodynamic therapy significantly suppresses the tumor growth and increases mouse survival compared with treatment with the NPs or TA99. The study reveals a new avenue to treat cancer by NP hitchhiking of immune systems to enhance delivery of therapeutics into tumor sites.
Collapse
Affiliation(s)
- Dafeng Chu
- Department of Pharmaceutical Sciences; College of Pharmacy; Washington State University; Spokane WA 99210 USA
| | - Qi Zhao
- Faculty of Health Sciences; University of Macau; Macau China
| | - Jian Yu
- Moores Cancer Center; University of California; San Diego La Jolla CA 92093 USA
| | - Faya Zhang
- Department of Pharmaceutical Sciences; College of Pharmacy; Washington State University; Spokane WA 99210 USA
| | - Hui Zhang
- Department of Pharmaceutical Sciences; College of Pharmacy; Washington State University; Spokane WA 99210 USA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences; College of Pharmacy; Washington State University; Spokane WA 99210 USA
| |
Collapse
|
19
|
Abstract
The capacity of immunoglobulin G (IgG) antibodies to eliminate virtually any target cell has resulted in the widespread introduction of cytotoxic antibodies into the clinic in settings of cancer therapy, autoimmunity, and transplantation, for example. More recently, it has become apparent that also the protection from viral infection via IgG antibodies may require cytotoxic effector functions, suggesting that antibody-dependent cellular cytotoxicity (ADCC) directed against malignant or virally infected cells is one of the most essential effector mechanisms triggered by IgG antibodies to protect the host. A detailed understanding of the underlying molecular and cellular pathways is critical, therefore, to make full use of this antibody effector function. Several studies over the last years have provided novel insights into the effector pathways and innate immune effector cells responsible for ADCC reactions. One of the most notable outcomes of many of these reports is that cells of the mononuclear phagocytic system rather than natural killer cells are critical for removal of IgG opsonized target cells in vivo.
Collapse
|
20
|
Ya Z, Hailemichael Y, Overwijk W, Restifo NP. Mouse model for pre-clinical study of human cancer immunotherapy. CURRENT PROTOCOLS IN IMMUNOLOGY 2015; 108:20.1.1-20.1.43. [PMID: 25640991 PMCID: PMC4361407 DOI: 10.1002/0471142735.im2001s108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This unit describes protocols for developing tumors in mice, including subcutaneous growth, pulmonary metastases of B16 melanoma, and spontaneous melanoma in B-Raf V600E/PTEN deletion transgenic mouse models. Two immunization methods to prevent B16 tumor growth are described using B16.GM-CSF and recombinant vaccinia virus. A therapeutic approach is also included that uses adoptive transfer of tumor antigen-specific T cells. Methods including CTL induction, isolation, testing, and genetic modification of mouse T cells for adoptive transfer by using retrovirus-expressing genes of interest are provided. Additional sections, including growing B16 melanoma, enumerating pulmonary metastases, tumor imaging technique, and use of recombinant viruses for vaccination, are discussed together with safety concerns.
Collapse
MESH Headings
- Animals
- Antibodies/blood
- Antibodies/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/immunology
- Cell Culture Techniques
- Cell- and Tissue-Based Therapy/adverse effects
- Cell- and Tissue-Based Therapy/methods
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Transfer Techniques
- Genetic Vectors/genetics
- Immunization/methods
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Male
- Melanoma, Experimental/diagnosis
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Transgenic
- Molecular Imaging/methods
- Neoplasm Metastasis
- Neoplasms/diagnosis
- Neoplasms/etiology
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transduction, Genetic
- Translational Research, Biomedical
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Zhiya Ya
- National Cancer Institute, Surgery Branch, Bethesda, Maryland
| | - Yared Hailemichael
- Department of Melanoma Medical Oncology-Research, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Willem Overwijk
- Department of Melanoma Medical Oncology-Research, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
21
|
Moskalenko M, Pan M, Fu Y, de Moll EH, Hashimoto D, Mortha A, Leboeuf M, Jayaraman P, Bernardo S, Sikora AG, Wolchok J, Bhardwaj N, Merad M, Saenger Y. Requirement for innate immunity and CD90⁺ NK1.1⁻ lymphocytes to treat established melanoma with chemo-immunotherapy. Cancer Immunol Res 2015; 3:296-304. [PMID: 25600438 DOI: 10.1158/2326-6066.cir-14-0120] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We sought to define cellular immune mechanisms of synergy between tumor-antigen-targeted monoclonal antibodies and chemotherapy. Established B16 melanoma in mice was treated with cytotoxic doses of cyclophosphamide in combination with an antibody targeting tyrosinase-related protein 1 (αTRP1), a native melanoma differentiation antigen. We find that Fcγ receptors are required for efficacy, showing that antitumor activity of combination therapy is immune mediated. Rag1(-/-) mice deficient in adaptive immunity are able to clear tumors, and thus innate immunity is sufficient for efficacy. Furthermore, previously treated wild-type mice are not significantly protected against tumor reinduction, as compared with mice inoculated with irradiated B16 alone, consistent with a primarily innate immune mechanism of action of chemo-immunotherapy. In contrast, mice deficient in both classical natural killer (NK) lymphocytes and nonclassical innate lymphocytes (ILC) due to deletion of the IL2 receptor common gamma chain IL2γc(-/-)) are refractory to chemo-immunotherapy. Classical NK lymphocytes are not critical for treatment, as depletion of NK1.1⁺ cells does not impair antitumor effect. Depletion of CD90⁺NK1.1⁻ lymphocytes, however, both diminishes therapeutic benefit and decreases accumulation of macrophages within the tumor. Tumor clearance during combination chemo-immunotherapy with monoclonal antibodies against native antigen is mediated by the innate immune system. We highlight a novel potential role for CD90⁺NK1.1⁻ ILCs in chemo-immunotherapy.
Collapse
Affiliation(s)
- Marina Moskalenko
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Michael Pan
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yichun Fu
- Department of Medicine, Columbia University Medical Center, New York, New York
| | - Ellen H de Moll
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Daigo Hashimoto
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York. Department of Hematology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Arthur Mortha
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marylene Leboeuf
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Padmini Jayaraman
- Department of Otolaryngology, Baylor College of Medicine, Houston, Texas
| | - Sebastian Bernardo
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Andrew G Sikora
- Department of Otolaryngology, Baylor College of Medicine, Houston, Texas
| | - Jedd Wolchok
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nina Bhardwaj
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Miriam Merad
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yvonne Saenger
- Division of Hematology and Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York. Department of Medicine, Columbia University Medical Center, New York, New York. Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
22
|
Boross P, Jansen JHM, van Tetering G, Nederend M, Brandsma A, Meyer S, Torfs E, van den Ham HJ, Meulenbroek L, de Haij S, Leusen JHW. Anti-tumor activity of human IgG1 anti-gp75 TA99 mAb against B16F10 melanoma in human FcgammaRI transgenic mice. Immunol Lett 2014; 160:151-7. [PMID: 24613852 DOI: 10.1016/j.imlet.2014.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/06/2014] [Accepted: 02/07/2014] [Indexed: 12/17/2022]
Abstract
Patients suffering from advanced melanoma have a very poor prognosis. Despite recent advances in the understanding of oncogenic mechanisms and therapeutic interventions, the median survival of patients with metastatic disease is less than 12 months. Immunotherapy of melanoma has been intensely investigated and holds great promises. Tyrosinase-related protein-1 or gp75 (TYRP-1/gp75) antigen is a melanosomal polypeptide. It is the most abundant glycoprotein synthesized by pigmented melanocytes and melanomas. It is specific for melanocytes and both primary and metastatic melanomas. In mice, administration of the mouse mAb anti-gp75 TA99 prevents outgrowth of B16F10 melanoma metastases. The activity of TA99 is dependent on the presence and activity of the IgG specific, Fc receptors. TA99 cross-reacts with human gp75, and is currently being used for diagnosis of patients. Here, we sequenced mIgG2a TA99 and found that the locus harboring the endogenous light chain of the fusion partner in the TA99 hybridoma cells is not inactivated, resulting in the production of a mixed pool of mAbs that mitigates binding to gp75. Since human IgG1 (hIgG1) is the most frequently used mAb format in clinical studies, we produced a recombinant hIgG1 TA99 molecule. Whereas it is known that hIgG1 can functionally interact with mouse Fc receptors, we found that hIgG1 TA99 did not exhibit in vivo activity against B16F10 melanoma in wild type C57BL/6 mice. However, results obtained in this study demonstrated anti-tumor activity of hIgG1 TA99 in FcγRIIB knockout mice and in human FcγRI transgenic mice. These results emphasize the need for testing hIgG mAb in mice with functional human FcγRs.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacology
- Cross Reactions
- Humans
- Hybridomas/chemistry
- Hybridomas/immunology
- Immunoglobulin G/chemistry
- Immunoglobulin G/genetics
- Immunoglobulin G/pharmacology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Transgenic
- Molecular Sequence Data
- Oxidoreductases/antagonists & inhibitors
- Oxidoreductases/genetics
- Oxidoreductases/immunology
- Receptors, IgG/deficiency
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Recombinant Fusion Proteins/chemistry
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/pharmacology
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/therapy
- Transgenes
Collapse
Affiliation(s)
- Peter Boross
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - J H Marco Jansen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Geert van Tetering
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Maaike Nederend
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Arianne Brandsma
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Saskia Meyer
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Ellen Torfs
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Henk-Jan van den Ham
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Laura Meulenbroek
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Simone de Haij
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands
| | - Jeanette H W Leusen
- Immunotherapy Laboratory, Laboratory for Translational Immunology, University Medical Center Utrecht, The Netherlands.
| |
Collapse
|
23
|
Scheiermann J, Klinman DM. Clinical evaluation of CpG oligonucleotides as adjuvants for vaccines targeting infectious diseases and cancer. Vaccine 2014; 32:6377-89. [PMID: 24975812 DOI: 10.1016/j.vaccine.2014.06.065] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/28/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022]
Abstract
Synthetic oligonucleotides (ODN) that express unmethylated "CpG motifs" trigger cells that express Toll-like receptor 9. In humans this includes plasmacytoid dendritic cells and B cells. CpG ODN induce an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. Their utility as vaccine adjuvants was evaluated in a number of clinical trials. Results indicate that CpG ODN improve antigen presentation and the generation of vaccine-specific cellular and humoral responses. This work provides an up-to-date overview of the utility of CpG ODN as adjuvants for vaccines targeting infectious agents and cancer.
Collapse
Affiliation(s)
- Julia Scheiermann
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, United States
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, United States.
| |
Collapse
|
24
|
Johnson AK. Repercussions of occult malignancy – An etiologic basis for rheumatic disease. Med Hypotheses 2013; 80:447-51. [DOI: 10.1016/j.mehy.2012.12.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 12/29/2012] [Indexed: 11/30/2022]
|
25
|
Schweighoffer T. Molecular cancer vaccines: Tumor therapy using antigen-specific immunizations. Pathol Oncol Res 2012; 3:164-76. [PMID: 18470726 DOI: 10.1007/bf02899917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/1997] [Accepted: 08/24/1997] [Indexed: 10/21/2022]
Abstract
Vaccination against tumors promises selective destruction of malignant cells by the host's immune system. Molecular cancer vaccines rely on recently identified tumor antigens as immunogens. Tumor antigens can be applied in many forms, as genes in recombinant vectors, as proteins or peptides representing T cell epitopes.Analysis of various aspects indicates some advantage for peptide-based vaccines over the other modalities. Further refinements and extensively monitored clinical trials are necessary to advance molecular cancer vaccines from concepts into powerful therapy.
Collapse
Affiliation(s)
- T Schweighoffer
- Department Cell Biology, Boehringer Ingelheim Research and Development, Dr. Boehringer-Gasse 5, A-l 120, Wien, Austria,
| |
Collapse
|
26
|
Noguchi T, Kato T, Wang L, Maeda Y, Ikeda H, Sato E, Knuth A, Gnjatic S, Ritter G, Sakaguchi S, Old LJ, Shiku H, Nishikawa H. Intracellular Tumor-Associated Antigens Represent Effective Targets for Passive Immunotherapy. Cancer Res 2012; 72:1672-82. [DOI: 10.1158/0008-5472.can-11-3072] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Monoclonal antibody (mAb) therapy against tumor antigens expressed on the tumor surface is associated with clinical benefit. However, many tumor antigens are intracellular molecules that generally would not be considered suitable targets for mAb therapy. In this study, we provide evidence challenging this view through an investigation of the efficacy of mAb directed against NY-ESO-1, a widely expressed immunogen in human tumors that is expressed intracellularly rather than on the surface of cells. On their own, NY-ESO-1 mAb could neither augment antigen-specific CD8+ T-cell induction nor cause tumor eradication. To facilitate mAb access to intracellular target molecules, we combined anti-NY-ESO-1 mAb with anticancer drugs to accentuate the release of intracellular NY-ESO-1 from dying tumor cells. Strikingly, combination therapy induced a strong antitumor effect that was accompanied by the development of NY-ESO-1–specific effector/memory CD8+ T cells that were not elicited by single treatments alone. The combinatorial effect was also associated with upregulation of maturation markers on dendritic cells, consistent with the organization of an effective antitumor T-cell response. Administration of Fc-depleted F(ab) mAb or combination treatment in Fcγ receptor–deficient host mice abolished the therapeutic effect. Together, our findings show that intracellular tumor antigens can be captured by mAbs and engaged in an efficient induction of CD8+ T-cell responses, greatly expanding the possible use of mAb for passive cancer immunotherapy. Cancer Res; 72(7); 1672–82. ©2012 AACR.
Collapse
Affiliation(s)
- Takuro Noguchi
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Takuma Kato
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Linan Wang
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Yuka Maeda
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Hiroaki Ikeda
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Eiichi Sato
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Alexander Knuth
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Sacha Gnjatic
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Gerd Ritter
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Shimon Sakaguchi
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Lloyd J. Old
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Hiroshi Shiku
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| | - Hiroyoshi Nishikawa
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
- Authors' Affiliations: Departments of 1Cancer Vaccine, 2Cellular and Molecular Immunology, and 3Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie; 4Department of Surgical Oncology, Hokkaido University Graduate School of Medicine, Hokkaido; 5Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka; 6Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan; 7Department of Oncology, University Hospital Zurich, Zurich, Switzerland; and 8Ludwig Institute for Cancer Research, New York Branch, Memorial Sloan-Kettering Cancer Center, New York
| |
Collapse
|
27
|
A targeted complement-dependent strategy to improve the outcome of mAb therapy, and characterization in a murine model of metastatic cancer. Blood 2012; 119:6043-51. [PMID: 22442351 DOI: 10.1182/blood-2011-10-383232] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Complement inhibitors expressed on tumor cells provide an evasion mechanism against mAb therapy and may modulate the development of an acquired antitumor immune response. Here we investigate a strategy to amplify mAb-targeted complement activation on a tumor cell, independent of a requirement to target and block complement inhibitor expression or function, which is difficult to achieve in vivo. We constructed a murine fusion protein, CR2Fc, and demonstrated that the protein targets to C3 activation products deposited on a tumor cell by a specific mAb, and amplifies mAb-dependent complement activation and tumor cell lysis in vitro. In syngeneic models of metastatic lymphoma (EL4) and melanoma (B16), CR2Fc significantly enhanced the outcome of mAb therapy. Subsequent studies using the EL4 model with various genetically modified mice and macrophage-depleted mice revealed that CR2Fc enhanced the therapeutic effect of mAb therapy via both macrophage-dependent FcγR-mediated antibody-dependent cellular cytotoxicity, and by direct complement-mediated lysis. Complement activation products can also modulate adaptive immunity, but we found no evidence that either mAb or CR2Fc treatment had any effect on an antitumor humoral or cellular immune response. CR2Fc represents a potential adjuvant treatment to increase the effectiveness of mAb therapy of cancer.
Collapse
|
28
|
Lesokhin AM, Hohl TM, Kitano S, Cortez C, Hirschhorn-Cymerman D, Avogadri F, Rizzuto GA, Lazarus JJ, Pamer EG, Houghton AN, Merghoub T, Wolchok JD. Monocytic CCR2(+) myeloid-derived suppressor cells promote immune escape by limiting activated CD8 T-cell infiltration into the tumor microenvironment. Cancer Res 2011; 72:876-86. [PMID: 22174368 DOI: 10.1158/0008-5472.can-11-1792] [Citation(s) in RCA: 297] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of cells that accumulate during tumor formation, facilitate immune escape, and enable tumor progression. MDSCs are important contributors to the development of an immunosuppressive tumor microenvironment that blocks the action of cytotoxic antitumor T effector cells. Heterogeneity in these cells poses a significant barrier to studying the in vivo contributions of individual MDSC subtypes. Herein, we show that granulocyte-macrophage colony stimulating factor, a cytokine critical for the numeric and functional development of MDSC populations, promotes expansion of a monocyte-derived MDSC population characterized by expression of CD11b and the chemokine receptor CCR2. Using a toxin-mediated ablation strategy to target CCR2-expressing cells, we show that these monocytic MDSCs regulate entry of activated CD8 T cells into the tumor site, thereby limiting the efficacy of immunotherapy. Our results argue that therapeutic targeting of monocytic MDSCs would enhance outcomes in immunotherapy.
Collapse
|
29
|
CD47-signal regulatory protein-α (SIRPα) interactions form a barrier for antibody-mediated tumor cell destruction. Proc Natl Acad Sci U S A 2011; 108:18342-7. [PMID: 22042861 DOI: 10.1073/pnas.1106550108] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Monoclonal antibodies are among the most promising therapeutic agents for treating cancer. Therapeutic cancer antibodies bind to tumor cells, turning them into targets for immune-mediated destruction. We show here that this antibody-mediated killing of tumor cells is limited by a mechanism involving the interaction between tumor cell-expressed CD47 and the inhibitory receptor signal regulatory protein-α (SIRPα) on myeloid cells. Mice that lack the SIRPα cytoplasmic tail, and hence its inhibitory signaling, display increased antibody-mediated elimination of melanoma cells in vivo. Moreover, interference with CD47-SIRPα interactions by CD47 knockdown or by antagonistic antibodies against CD47 or SIRPα significantly enhances the in vitro killing of trastuzumab-opsonized Her2/Neu-positive breast cancer cells by phagocytes. Finally, the response to trastuzumab therapy in breast cancer patients appears correlated to cancer cell CD47 expression. These findings demonstrate that CD47-SIRPα interactions participate in a homeostatic mechanism that restricts antibody-mediated killing of tumor cells. This provides a rational basis for targeting CD47-SIRPα interactions, using for instance the antagonistic antibodies against human SIRPα described herein, to potentiate the clinical effects of cancer therapeutic antibodies.
Collapse
|
30
|
The Association of Intratumoral Germinal Centers with Early-Stage Non-small Cell Lung Cancer. J Thorac Oncol 2011; 6:1687-90. [DOI: 10.1097/jto.0b013e3182217bec] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
31
|
Byrne KT, Turk MJ. New perspectives on the role of vitiligo in immune responses to melanoma. Oncotarget 2011; 2:684-94. [PMID: 21911918 PMCID: PMC3248219 DOI: 10.18632/oncotarget.323] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 09/09/2011] [Indexed: 01/09/2023] Open
Abstract
Melanoma-associated vitiligo is the best-studied example of the linkage between tumor immunity and autoimmunity. Although vitiligo is an independent positive prognostic factor for melanoma patients, the autoimmune destruction of melanocytes was long thought to be merely a side effect of robust anti-tumor immunity. However, new data reveal a key role for vitiligo in supporting T cell responses to melanoma. This research perspective reviews the history of melanoma-associated vitiligo in patients, the experimental studies that form the basis for understanding this relationship, and the unique characteristics of melanoma-specific CD8 T cells found in hosts with vitiligo. We also discuss the implications of our recent findings for the interpretation of patient responses, and the design of next-generation cancer immunotherapies.
Collapse
Affiliation(s)
- Katelyn T Byrne
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, NH, USA
| | | |
Collapse
|
32
|
Kim SK, Wu X, Ragupathi G, Gathuru J, Koide F, Cheung NK, Panageas K, Livingston PO. Impact of minimal tumor burden on antibody response to vaccination. Cancer Immunol Immunother 2011; 60:621-7. [PMID: 21267719 PMCID: PMC3734789 DOI: 10.1007/s00262-011-0975-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 01/05/2011] [Indexed: 10/18/2022]
Abstract
Four randomized phase III trials conducted recently in melanoma patients in the adjuvant setting have been based in part on the correlation between antibody responses in immunized patients and improved survival. Each of these randomized trials demonstrated no clinical benefit, although again there was a significant correlation between antibody response after vaccination and disease free and overall survival. To better understand this paradox, we established a surgical adjuvant model targeting GD2 ganglioside on EL4 lymphoma cells injected into the foot pad followed by amputation at variable intervals. Our findings are (1) comparable strong therapeutic benefit resulted from treatment of mice after amputation with a GD2-KLH conjugate vaccine or with anti-GD2 monoclonal antibody 3F8. (2) The strongest correlation was between antibody induction in response to vaccination and prolonged survival. (3) Antibody titers in response to vaccination in tumor challenged mice as compared to unchallenged mice were far lower despite the absence of detectable recurrences at the time. (4) The half life of administered 3F8 monoclonal antibody (but not control antibody) in challenged mice administered was significantly shorter than the half life of 3F8 antibody in unchallenged controls. The correlation between vaccine-induced antibody titers and prolonged survival may reflect, at least in part, increased tumor burden in antibody-negative mice. Absorption of vaccine-induced antibodies by increased, although not detected tumor burden may also explain the correlation between vaccine-induced antibody titers and survival in the adjuvant clinical trials described above.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Murine-Derived
- Antibodies, Neoplasm/biosynthesis
- Antibody-Dependent Cell Cytotoxicity
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Disease-Free Survival
- Enzyme-Linked Immunosorbent Assay
- Gangliosides/immunology
- Hemocyanins/immunology
- Immunoglobulin G/immunology
- Immunoglobulin G/therapeutic use
- Lymphoma/immunology
- Lymphoma/pathology
- Lymphoma/therapy
- Mice
- Mice, Inbred C57BL
- Tumor Burden
- Vaccination
- Vaccines, Conjugate/administration & dosage
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/therapeutic use
Collapse
Affiliation(s)
| | - Xiaohong Wu
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Govind Ragupathi
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | | | - Nai-Kong Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Katherine Panageas
- Department of Biostatistics, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | - Philip O. Livingston
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Only a subset of melanoma patients with advanced disease seems to benefit from immunotherapy. Predictive markers identifying these patients are unfortunately not available. Whether immune-related side effects could serve as predictors for treatment response or just resemble unwanted side effects from immunotherapy will be outlined in this review. RECENT FINDINGS Early studies suggested an association of immune-related side effects such as vitiligo and autoimmune thyroiditis with response in patients receiving IL-2 or IFNα. However, conflicting data have been reported as well, mentioning the effect of a higher rate of immune-related toxicities during prolonged administration of the drug in responders/survivors. This type of bias is also known as guarantee-time bias. Recently, a clearly significant and clinically relevant prolongation of survival was demonstrated in patients with metastatic melanoma treated with ipilimumab. Immune-related adverse events were associated with response to ipilimumab, however, at the cost of considerable toxicity. SUMMARY Evidence for an association of immune-related toxicities and response in patients receiving IL-2 or IFNα is weak, considering guarantee-time bias. On the contrary, this association for patients receiving anti-cytotoxic T-lymphocyte antigen-4 therapy (ipilimumab) appears much stronger. Importantly, can we uncouple tumor immunity from autoimmunity in order to optimize immunotherapy in melanoma?
Collapse
|
34
|
Bouwhuis MG, ten Hagen TLM, Eggermont AMM. Immunologic functions as prognostic indicators in melanoma. Mol Oncol 2011; 5:183-9. [PMID: 21367679 DOI: 10.1016/j.molonc.2011.01.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 01/27/2011] [Indexed: 02/08/2023] Open
Abstract
Outcome in melanoma patients with advanced disease is poor and systemic treatment seems to benefit only a subset of patients. Predictive markers identifying these patients are currently not available. Early studies showed an association of immune-related side effects such as vitiligo and autoimmune thyroiditis with response to IL-2 or IFNα treatment. However, conflicting data have been reported as well, mentioning the effect of a higher rate of immune-related toxicities during prolonged administration of the drug in responders. The review discusses the prognostic significance of autoimmunity during various forms of immunotherapy and stresses the importance of correcting for guarantee-time bias. In addition, other immune-related factors which have been associated with melanoma prognosis such as, CRP, white blood cell count, absolute lymphocyte count and human leukocyte antigen will be reviewed as well. A better understanding of the immune system and the host-tumor interactions should ultimately lead to more effective treatment. A major challenge expected to be addressed in future is proving ways to uncouple tumor immunity from autoimmunity.
Collapse
Affiliation(s)
- Marna G Bouwhuis
- Department of Surgery, Division Surgical Oncology, Erasmus University Medical Center - Daniel den Hoed Cancer Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
35
|
Tyrosinase related protein 1 (TYRP1/gp75) in human cutaneous melanoma. Mol Oncol 2011; 5:150-5. [PMID: 21324755 DOI: 10.1016/j.molonc.2011.01.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2010] [Accepted: 01/27/2011] [Indexed: 02/06/2023] Open
Abstract
Melanoma prognosis is based on specific pathological features at the primary lesion. In metastatic patients, the extent of lymph node involvement is also an important prognosis indicator. Many progression markers both in tissues and serum, including circulating tumor cells, have been studied and new molecular markers are awaited from high-throughput screenings to discriminate between clinical stages and predict disease progression. The present review focuses on human tyrosinase related protein 1 also known as gp75 glycoprotein (Tyrp1/gp75), a melanosomal protein involved in the pigmentary machinery of the melanocyte and often used as differentiation marker, with a special emphasis on its emerging roles in the malignant melanocyte and melanoma progression.
Collapse
|
36
|
Alphavirus replicon particles expressing TRP-2 provide potent therapeutic effect on melanoma through activation of humoral and cellular immunity. PLoS One 2010; 5. [PMID: 20844763 PMCID: PMC2937034 DOI: 10.1371/journal.pone.0012670] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 08/16/2010] [Indexed: 02/06/2023] Open
Abstract
Background Malignant melanoma is the deadliest form of skin cancer and is refractory to conventional chemotherapy and radiotherapy. Therefore alternative approaches to treat this disease, such as immunotherapy, are needed. Melanoma vaccine design has mainly focused on targeting CD8+ T cells. Activation of effector CD8+ T cells has been achieved in patients, but provided limited clinical benefit, due to immune-escape mechanisms established by advanced tumors. We have previously shown that alphavirus-based virus-like replicon particles (VRP) simultaneously activate strong cellular and humoral immunity against the weakly immunogenic melanoma differentiation antigen (MDA) tyrosinase. Here we further investigate the antitumor effect and the immune mechanisms of VRP encoding different MDAs. Methodology/Principal Findings VRP encoding different MDAs were screened for their ability to prevent the growth of the B16 mouse transplantable melanoma. The immunologic mechanisms of efficacy were investigated for the most effective vaccine identified, focusing on CD8+ T cells and humoral responses. To this end, ex vivo immune assays and transgenic mice lacking specific immune effector functions were used. The studies identified a potent therapeutic VRP vaccine, encoding tyrosinase related protein 2 (TRP-2), which provided a durable anti-tumor effect. The efficacy of VRP-TRP2 relies on a novel immune mechanism of action requiring the activation of both IgG and CD8+ T cell effector responses, and depends on signaling through activating Fcγ receptors. Conclusions/Significance This study identifies a VRP-based vaccine able to elicit humoral immunity against TRP-2, which plays a role in melanoma immunotherapy and synergizes with tumor-specific CD8+ T cell responses. These findings will aid in the rational design of future immunotherapy clinical trials.
Collapse
|
37
|
Metastatic melanomas express inhibitory low affinity fc gamma receptor and escape humoral immunity. Dermatol Res Pract 2010; 2010:657406. [PMID: 20672001 PMCID: PMC2905727 DOI: 10.1155/2010/657406] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 04/01/2010] [Indexed: 12/22/2022] Open
Abstract
Our research, inspired by the pioneering works of Isaac Witz in the 1980s, established that 40% of human metastatic melanomas express ectopically inhibitory Fc gamma receptors (FcγRIIB), while they are detected on less than 5% of primary cutaneous melanoma and not on melanocytes. We demonstrated that these tumoral FcγRIIB act as decoy receptors that bind the Fc portion of antimelanoma IgG, which may prevent Fc recognition by the effector cells of the immune system and allow the metastatic melanoma to escape the humoral/natural immune response. The FcγRIIB is able to inhibit the ADCC (antibody dependent cell cytotoxicity) in vitro. Interestingly, the percentage of melanoma expressing the FcγRIIB is high (70%) in organs like the liver, which is rich in patrolling NK (natural killer) cells that exercise their antitumoral activity by ADCC. We found that this tumoral FcγRIIB is fully functional and that its inhibitory potential can be triggered depending on the specificity of the anti-tumor antibody with which it interacts.
Together these observations elucidate how metastatic melanomas interact with and potentially evade humoral immunity and provide direction for the improvement of anti-melanoma monoclonal antibody therapy.
Collapse
|
38
|
Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, Kim SC, Diab A, Rizzuto G, Duan F, Perales MA, Merghoub T, Houghton AN, Wolchok JD. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS One 2010; 5:e10436. [PMID: 20454651 PMCID: PMC2862699 DOI: 10.1371/journal.pone.0010436] [Citation(s) in RCA: 196] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 04/08/2010] [Indexed: 11/19/2022] Open
Abstract
In vivo GITR ligation has previously been shown to augment T-cell-mediated anti-tumor immunity, yet the underlying mechanisms of this activity, particularly its in vivo effects on CD4+ foxp3+ regulatory T cells (Tregs), have not been fully elucidated. In order to translate this immunotherapeutic approach to the clinic it is important gain better understanding of its mechanism(s) of action. Utilizing the agonist anti-GITR monoclonal antibody DTA-1, we found that in vivo GITR ligation modulates regulatory T cells (Tregs) directly during induction of melanoma tumor immunity. As a monotherapy, DTA-1 induced regression of small established B16 melanoma tumors. Although DTA-1 did not alter systemic Treg frequencies nor abrogate the intrinsic suppressive activity of Tregs within the tumor-draining lymph node, intra-tumor Treg accumulation was significantly impaired. This resulted in a greater Teff:Treg ratio and enhanced tumor-specific CD8+ T-cell activity. The decreased intra-tumor Treg accumulation was due both to impaired infiltration, coupled with DTA-1-induced loss of foxp3 expression in intra-tumor Tregs. Histological analysis of B16 tumors grown in Foxp3-GFP mice showed that the majority of GFP+ cells had lost Foxp3 expression. These "unstable" Tregs were absent in IgG-treated tumors and in DTA-1 treated TDLN, demonstrating a tumor-specific effect. Impairment of Treg infiltration was lost if Tregs were GITR(-/-), and the protective effects of DTA-1 were reduced in reconstituted RAG1(-/-) mice if either the Treg or Teff subset were GITR-negative and absent if both were negative. Our results demonstrate that DTA-1 modulates both Teffs and Tregs during effective tumor treatment. The data suggest that DTA-1 prevents intra-tumor Treg accumulation by altering their stability, and as a result of the loss of foxp3 expression, may modify their intra-tumor suppressive capacity. These findings provide further support for the continued development of agonist anti-GITR mAbs as an immunotherapeutic strategy for cancer.
Collapse
Affiliation(s)
- Adam D. Cohen
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - David A. Schaer
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Cailian Liu
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Yanyun Li
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Daniel Hirschhorn-Cymmerman
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Soo Chong Kim
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Adi Diab
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Gabrielle Rizzuto
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - Fei Duan
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - Miguel A. Perales
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - Taha Merghoub
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Alan N. Houghton
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - Jedd D. Wolchok
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
39
|
Cohen AD, Schaer DA, Liu C, Li Y, Hirschhorn-Cymmerman D, Kim SC, Diab A, Rizzuto G, Duan F, Perales MA, Merghoub T, Houghton AN, Wolchok JD. Agonist anti-GITR monoclonal antibody induces melanoma tumor immunity in mice by altering regulatory T cell stability and intra-tumor accumulation. PLoS One 2010. [PMID: 20454651 DOI: 10.1371/journal.pone.0010436.s007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In vivo GITR ligation has previously been shown to augment T-cell-mediated anti-tumor immunity, yet the underlying mechanisms of this activity, particularly its in vivo effects on CD4+ foxp3+ regulatory T cells (Tregs), have not been fully elucidated. In order to translate this immunotherapeutic approach to the clinic it is important gain better understanding of its mechanism(s) of action. Utilizing the agonist anti-GITR monoclonal antibody DTA-1, we found that in vivo GITR ligation modulates regulatory T cells (Tregs) directly during induction of melanoma tumor immunity. As a monotherapy, DTA-1 induced regression of small established B16 melanoma tumors. Although DTA-1 did not alter systemic Treg frequencies nor abrogate the intrinsic suppressive activity of Tregs within the tumor-draining lymph node, intra-tumor Treg accumulation was significantly impaired. This resulted in a greater Teff:Treg ratio and enhanced tumor-specific CD8+ T-cell activity. The decreased intra-tumor Treg accumulation was due both to impaired infiltration, coupled with DTA-1-induced loss of foxp3 expression in intra-tumor Tregs. Histological analysis of B16 tumors grown in Foxp3-GFP mice showed that the majority of GFP+ cells had lost Foxp3 expression. These "unstable" Tregs were absent in IgG-treated tumors and in DTA-1 treated TDLN, demonstrating a tumor-specific effect. Impairment of Treg infiltration was lost if Tregs were GITR(-/-), and the protective effects of DTA-1 were reduced in reconstituted RAG1(-/-) mice if either the Treg or Teff subset were GITR-negative and absent if both were negative. Our results demonstrate that DTA-1 modulates both Teffs and Tregs during effective tumor treatment. The data suggest that DTA-1 prevents intra-tumor Treg accumulation by altering their stability, and as a result of the loss of foxp3 expression, may modify their intra-tumor suppressive capacity. These findings provide further support for the continued development of agonist anti-GITR mAbs as an immunotherapeutic strategy for cancer.
Collapse
Affiliation(s)
- Adam D Cohen
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhou P, Fang X, McNally BA, Yu P, Zhu M, Fu YX, Wang L, Liu Y, Zheng P. Targeting lymphotoxin-mediated negative selection to prevent prostate cancer in mice with genetic predisposition. Proc Natl Acad Sci U S A 2009; 106:17134-9. [PMID: 19805094 PMCID: PMC2761305 DOI: 10.1073/pnas.0905707106] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Indexed: 02/01/2023] Open
Abstract
The identification of individuals genetically susceptible to cancer calls for preventive measures to minimize the cancer risk in these high-risk populations. Immune prevention is made necessary by the anticipated health threat, but lack of enough high-affinity T cells against tumor-associated antigens and the unpredictability of tumor antigens make antigen-based immune prevention untenable for cancer. To address this issue, we explored a non-antigen-based cancer immune prevention strategy using the transgenic adenocarcinoma of mouse prostate model that spontaneously develops prostate cancer with 100% penetrance. We show that targeted mutation of the lymphotoxin alpha (LTalpha) gene efficiently rescued tumor-reactive T cells, drastically reduced cancer incidence, and almost completely ablated metastasis. Remarkably, short-term treatments with the fusion protein consisting of constant region of IgG and extracellular domain of lymphotoxin beta receptor (LTbetaRIg) interrupted clonal deletion, reduced the size of the primary cancer, and completely prevented metastasis later in life. Our data demonstrated the value of non-antigen-based immune prevention for those with a genetic predisposition to cancer.
Collapse
Affiliation(s)
- Penghui Zhou
- Division of Immunotherapy, Departments of Surgery, Internal Medicine, and Pathology, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Xianfeng Fang
- Institute of Biophysics, Chinese Academy of Science, Beijing 100101, China; and
| | - Beth A. McNally
- Division of Immunotherapy, Departments of Surgery, Internal Medicine, and Pathology, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Ping Yu
- Department of Pathology, University of Chicago School of Medicine, Chicago, IL 60636
| | - Mingzhao Zhu
- Department of Pathology, University of Chicago School of Medicine, Chicago, IL 60636
| | - Yang-Xin Fu
- Department of Pathology, University of Chicago School of Medicine, Chicago, IL 60636
| | - Lizhong Wang
- Division of Immunotherapy, Departments of Surgery, Internal Medicine, and Pathology, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Yang Liu
- Division of Immunotherapy, Departments of Surgery, Internal Medicine, and Pathology, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| | - Pan Zheng
- Division of Immunotherapy, Departments of Surgery, Internal Medicine, and Pathology, University of Michigan School of Medicine and Cancer Center, Ann Arbor, MI 48109
| |
Collapse
|
41
|
Clark AJ, Diamond M, Elfline M, Petty HR. Calicum microdomains form within neutrophils at the neutrophil-tumor cell synapse: role in antibody-dependent target cell apoptosis. Cancer Immunol Immunother 2009; 59:149-59. [PMID: 19593564 DOI: 10.1007/s00262-009-0735-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 06/29/2009] [Indexed: 01/03/2023]
Abstract
Ca(2+) messages are broadly important in cellular signal transduction. In immune cells, Ca(2+) signaling is an essential step in many forms of activation. Neutrophil-mediated antibody-dependent cell-mediated cytotoxicity (ADCC) is one form of leukocyte activation that plays an important role in tumor cell killing in vitro and in patient care. Using fluorescence methodologies, we found that neutrophils exhibit Ca(2+) signals during ADCC directed against breast fibrosarcoma cells. Importantly, these signals were localized to Ca(2+) microdomains at the neutrophil-to-tumor cell interface where they display dynamic features such as movement, fusion, and fission. These signals were blocked by the intracellular Ca(2+) buffer BAPTA. At the neutrophil-tumor cell synapse, the neutrophil's cytoplasm was enriched in STIM1, a crucial mediator of Ca(2+) signaling, whereas the Ca(2+)-binding proteins calbindin and parvalbumin were not affected. Our findings suggest that Ca(2+) microdomains are due to an active signaling process. As Ca(2+) signals within neutrophils were necessary for specific tumor cell apoptosis, a central role of microdomains in leukocyte-mediated tumor cell destruction is indicated.
Collapse
Affiliation(s)
- Andrea J Clark
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA
| | | | | | | |
Collapse
|
42
|
Oble DA, Loewe R, Yu P, Mihm MC. Focus on TILs: prognostic significance of tumor infiltrating lymphocytes in human melanoma. CANCER IMMUNITY 2009; 9:3. [PMID: 19338264 PMCID: PMC2935762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Tumors contain variable numbers of lymphocytes, referred to as tumor infiltrating lymphocytes (TILs). In melanoma, the intensity of this lymphocytic infiltrate is believed to correlate with outcome, though there is some debate about the applicability of this finding for all melanomas. Much research has gone into classifying TILs with respect to antigen receptor structure and the antigen to which melanoma-specific T cells react. However, these studies for the most part did not immunophenotype TILs, and recent data has revealed that the composition of tumoral lymphocytes is not homogenous, but rather represents varying contributions from many lymphocytic subsets. Furthermore, the function of TILs is often compromised as a result of the accumulation of immunoregulatory cells and various tumor escape mechanisms. These recent insights stress the need to collect more data on the composition and function of TIL infiltrates before definitive conclusions about the prognostic significance of TILs can be drawn. Advances in immunology have also facilitated the development of immunotherapeutic strategies, examples of which will be discussed with a special emphasis on blocking antibodies against CTLA-4, which are prototypical immunotherapeutic agents. This flurry of novel "biological" therapies will undoubtedly complicate our already incomplete understanding of TIL immunobiology as each of these agents has the potential to uniquely distort the series of immunological events which normally occur in untreated melanoma. Therefore, considerable research is needed to better elucidate the function and prognostic significance of TILs in both untreated melanoma and tumors treated with "biological" therapy.
Collapse
MESH Headings
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/therapeutic use
- Antigens, CD/immunology
- Antigens, CD/metabolism
- CTLA-4 Antigen
- Cytokines/immunology
- Cytokines/metabolism
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Immunologic Factors/immunology
- Immunologic Factors/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Melanoma/drug therapy
- Melanoma/immunology
- Melanoma/metabolism
- Prognosis
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Skin Neoplasms/drug therapy
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
Collapse
Affiliation(s)
- Darryl A. Oble
- Department of Pathology, Massachusetts
General Hospital, Harvard Medical School55
Fruit Street, Warren 827Boston, MA 02114USA
- Department of Medicine, Section of Dermatology,
University of Chicago5841 S. Maryland Avenue, MC
5067Chicago, IL 60637USA
| | - Robert Loewe
- Department of Pathology, Massachusetts
General Hospital, Harvard Medical School55
Fruit Street, Warren 827Boston, MA 02114USA
| | - Ping Yu
- Department of Medicine, Section of Dermatology,
University of Chicago5841 S. Maryland Avenue, MC
5067Chicago, IL 60637USA
| | - Martin C. Mihm
- Department of Pathology, Massachusetts
General Hospital, Harvard Medical School55
Fruit Street, Warren 827Boston, MA 02114USA
- Department of Dermatology, Massachusetts
General Hospital, Harvard Medical School50
Staniford Street, Suite 200Boston, MA 02114USA
| |
Collapse
|
43
|
Saenger YM, Li Y, Chiou KC, Chan B, Rizzuto G, Terzulli SL, Merghoub T, Houghton AN, Wolchok JD. Improved tumor immunity using anti-tyrosinase related protein-1 monoclonal antibody combined with DNA vaccines in murine melanoma. Cancer Res 2009; 68:9884-91. [PMID: 19047169 DOI: 10.1158/0008-5472.can-08-2233] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Passive immunization with monoclonal antibody TA99 targeting melanoma differentiation antigen tyrosinase-related protein-1 (Tyrp1; gp75) and active immunization with plasmid DNA encoding altered Tyrp1 both mediate tumor immunity in the B16 murine melanoma model. We report here that TA99 enhances Tyrp1 DNA vaccination in the treatment of B16 lung metastases, an effect mediated by immunologic mechanisms as Tyrp1 has no known role in regulating tumor growth. TA99 is shown to increase induction of anti-Tyrp1 CD8+T-cell responses to DNA vaccination against Tyrp1 as assessed by IFN-gamma ELISPOT assays. Immunohistochemistry studies reveal that TA99 localizes rapidly and specifically to B16 lung nodules. Augmentation of T-cell responses is dependent on the presence of tumor as well as on activating Fc receptors. Furthermore, TA99 enhances DNA vaccination against a distinct melanoma antigen, gp100(pmel17/silver locus), improving antitumor efficacy, augmenting systemic CD8+ T-cell responses to gp100, and increasing CD8+ T-cell infiltration at the tumor site. Epitope spreading was observed, with CD8+ T-cell responses generated to Tyrp1 peptide in mice receiving gp100 DNA vaccination in the presence of TA99. Finally, we show that TA99 improves therapeutic efficacy of DNA vaccination combined with adoptive T-cell transfer in treatment of established subcutaneous B16 melanoma. In conclusion, TA99 enhances DNA vaccination against both the target antigen Tyrp1 and a distinct melanoma antigen gp100 in an Fc receptor-dependent mechanism, consistent with enhanced cross-presentation of tumor-derived antigen. Monoclonal antibodies should be tested as vaccine adjuvants in the treatment of cancer.
Collapse
Affiliation(s)
- Yvonne M Saenger
- The Swim Across America Laboratory, Immunology Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center, and Weill Medical College of Cornell University, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Cassard L, Cohen-Solal JFG, Fournier EM, Camilleri-Broët S, Spatz A, Chouaïb S, Badoual C, Varin A, Fisson S, Duvillard P, Boix C, Loncar SM, Sastre-Garau X, Houghton AN, Avril MF, Gresser I, Fridman WH, Sautès-Fridman C. Selective expression of inhibitory Fcgamma receptor by metastatic melanoma impairs tumor susceptibility to IgG-dependent cellular response. Int J Cancer 2009; 123:2832-9. [PMID: 18798552 DOI: 10.1002/ijc.23870] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
During melanoma progression, patients develop anti-tumor immunity including the production of anti-tumor antibodies. Although the strategies developed by malignant cells to escape anti-tumor cellular immunity have been extensively investigated, little is known about tumor resistance to humoral immunity. The main effect of IgG antibodies is to activate the immune response by binding to host Fc gamma receptors (FcgammaR) expressed by immune cells. We previously reported in a limited study that some human metastatic melanoma cells ectopically express the FcgammaRIIB1, an inhibitory isoform of FcgammaR. By analyzing a large panel of different types of human primary and metastatic solid tumors, we report herein that expression of FcgammaRIIB is restricted to melanoma and is acquired during tumor progression. We show that FcgammaRIIB expression prevents the lysis of human metastatic melanoma cells by NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) in vitro, independently of the intracytoplasmic region of FcgammaRIIB. Using experimental mouse models, we demonstrate that expression of FcgammaRIIB protects B16F0 melanoma tumors from the ADCC induced by monoclonal and polyclonal anti-tumor IgG in vivo. Thus, our results identify FcgammaRIIB as a marker of human metastatic melanoma that impairs the tumor susceptibility to FcgammaR-dependent innate effector responses.
Collapse
Affiliation(s)
- Lydie Cassard
- INSERM, U872, Microenvironnement immunitaire et tumeurs, Equipe 13, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Nakamura A, Kubo T, Takai T. Fc receptor targeting in the treatment of allergy, autoimmune diseases and cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 640:220-33. [PMID: 19065795 DOI: 10.1007/978-0-387-09789-3_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fc receptors (FcRs) play an important role in the maintenance of an adequate activation threshold of various cells in antibody-mediated immune responses. Analyses of murine models show that the inhibitory FcR, FcyRIIB plays a pivotal role in the suppression of antibody-mediated allergy and autoimmunity. On the other hand, the activating-type FcRs are essential for the development of these diseases, suggesting that regulation of inhibitory or activating FcR is an ideal target for a therapeutic agent. Recent experimental or clinical studies also indicate that FcRs function as key receptors in the treatment with monoclonal antibodies (mAbs) therapy. This review summarizes FcR functions and highlights possible FcR-targeting therapies including mAb therapies for allergy, autoimmune diseases and cancer.
Collapse
Affiliation(s)
- Akira Nakamura
- Department of Experimental Immunology and CREST program of Japan Science and Technology Agency, Institute of Development, Aging and Cancer, Tohoku University, Seiryo 4-1, Sendai 980-8575, Japan.
| | | | | |
Collapse
|
46
|
Otten MA, van der Bij GJ, Verbeek SJ, Nimmerjahn F, Ravetch JV, Beelen RHJ, van de Winkel JGJ, van Egmond M. Experimental Antibody Therapy of Liver Metastases Reveals Functional Redundancy between FcγRI and FcγRIV. THE JOURNAL OF IMMUNOLOGY 2008; 181:6829-36. [DOI: 10.4049/jimmunol.181.10.6829] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
47
|
Overwijk WW, Restifo NP. B16 as a mouse model for human melanoma. ACTA ACUST UNITED AC 2008; Chapter 20:Unit 20.1. [PMID: 18432774 DOI: 10.1002/0471142735.im2001s39] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This unit details protocols for in vivo models of subcutaneous growth and pulmonary metastases of B16 melanoma. Therapeutic approaches include the use of B16.GM-CSF and rVVmTRP-1 to induce autoimmune vitiligo and tumor protection. The induction and use of gp 100-specific therapeutic cytotoxic T lymphocytes (CTL) are discussed. Methods are also included for CTL induction, isolation and testing, CTL maintenance, and adoptive transfer. Support protocols detail the testing of mouse sera for presence of MDA-specific antibodies by immunoblotting and ELISA, respectively. Additional sections, including growing B16 melanoma, enumerating pulmonary metastases, and use of recombinant viruses for vaccination, are discussed together with safety concerns.
Collapse
Affiliation(s)
- W W Overwijk
- National Cancer Institute, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
48
|
Felicetti P, Mennecozzi M, Barucca A, Montgomery S, Orlandi F, Manova K, Houghton AN, Gregor PD, Concetti A, Venanzi FM. Tumor endothelial marker 8 enhances tumor immunity in conjunction with immunization against differentiation Ag. Cytotherapy 2008; 9:23-34. [PMID: 18236207 DOI: 10.1080/14653240601048369] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND We have previously shown that xenogeneic DNA vaccines encoding rat neu and melanosomal differentiation Ag induce tumor immunity. Others have developed vaccines targeting tumor neovasculature. Tumor endothelial marker 8 (TEM8) is expressed in the neovasculature of human tumors, and in the mouse melanoma B16, but its expression is limited in normal adult tissues. We describe a DNA vaccine combining xenogeneic tumor Ag and TEM8. METHODS In-situ hybridization was used to detect TEM8 RNA in mouse tumors. Mice transgenic for the rat neu proto-oncogene were immunized with DNA vaccines encoding TEM8 and the extracellular domain of rat neu and challenged with the 233-VSGA1 breast cancer cell line. In parallel experiments, C57BL/6 mice were immunized with TEM8 and human tyrosinase-related protein 1 (hTYRP1/hgp75) and challenged with B16F10 melanoma. RESULTS TEM8 was expressed in the stroma of transplantable mouse breast and melanoma tumors. In both model systems, TEM8 DNA had no activity as a single agent but significantly enhanced the anti-tumor immunity of neu and hTYRP1/hgp75 DNA vaccines when given in concert. The observed synergy was dependent upon CD8+ T cells, as depletion of this cell population just prior to tumor challenge obviated the effect of the TEM8 vaccine in both tumor models. DISCUSSION A local immune response to TEM8 may increase inflammation or tumor necrosis within the tumor, resulting in improved Ag presentation of HER2/neu and hTYRP1/hgp75. Alternatively, TEM8 expression in host APC may alter T-cell interactions or homing. In this way, TEM8 may act more as an adjuvant than an immunologic target.
Collapse
Affiliation(s)
- P Felicetti
- Swim Across America Laboratory of Tumor Immunology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Juang CM, Hung CF, Yeh JY, Horng HC, Twu NF, Cheng MH, Wen KC, Yuan CC, Chao KC, Wu TC, Yen MS. Regulatory T cells: potential target in anticancer immunotherapy. Taiwan J Obstet Gynecol 2007; 46:215-21. [PMID: 17962099 DOI: 10.1016/s1028-4559(08)60023-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The concept of regulatory T cells was first described in the early 1970s, and regulatory T cells were called suppressive T cells at that time. Studies that followed have demonstrated that these suppressive T cells negatively regulated tumor immunity and contributed to tumor growth in mice. Despite the importance of these studies, there was extensive skepticism about the existence of these cells, and the concept of suppressive T cells left the center stage of immunologic research for decades. Interleukin-2 receptor alpha-chain, CD25, was first demonstrated in 1995 to serve as a phenotypic marker for CD4+ regulatory cells. Henceforth, research of regulatory T cells boomed. Regulatory T cells are involved in the pathogenesis of cancer, autoimmune disease, transplantation immunology, and immune tolerance in pregnancy. Recent evidence has demonstrated that regulatory T cell-mediated immunosuppression is one of the crucial tumor immune evasion mechanisms and the main obstacle of successful cancer immunotherapy. The mechanism and the potential clinical application of regulatory T cells in cancer immunotherapy are discussed.
Collapse
Affiliation(s)
- Chi-Mou Juang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Valmori D, Souleimanian NE, Tosello V, Bhardwaj N, Adams S, O'Neill D, Pavlick A, Escalon JB, Cruz CM, Angiulli A, Angiulli F, Mears G, Vogel SM, Pan L, Jungbluth AA, Hoffmann EW, Venhaus R, Ritter G, Old LJ, Ayyoub M. Vaccination with NY-ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming. Proc Natl Acad Sci U S A 2007; 104:8947-52. [PMID: 17517626 PMCID: PMC1885608 DOI: 10.1073/pnas.0703395104] [Citation(s) in RCA: 248] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Indexed: 01/05/2023] Open
Abstract
The use of recombinant tumor antigen proteins is a realistic approach for the development of generic cancer vaccines, but the potential of this type of vaccines to induce specific CD8(+) T cell responses, through in vivo cross-priming, has remained unclear. In this article, we report that repeated vaccination of cancer patients with recombinant NY-ESO-1 protein, Montanide ISA-51, and CpG ODN 7909, a potent stimulator of B cells and T helper type 1 (Th1)-type immunity, resulted in the early induction of specific integrated CD4(+) Th cells and antibody responses in most vaccinated patients, followed by the development of later CD8(+) T cell responses in a fraction of them. The correlation between antibody and T cell responses, together with the ability of vaccine-induced antibodies to promote in vitro cross-presentation of NY-ESO-1 by dendritic cells to vaccine-induced CD8(+) T cells, indicated that elicitation of NY-ESO-1-specific CD8(+) T cell responses by cross-priming in vivo was associated with the induction of adequate levels of specific antibodies. Together, our data provide clear evidence of in vivo cross-priming of specific cytotoxic T lymphocytes by a recombinant tumor antigen vaccine, underline the importance of specific antibody induction for the cross-priming to occur, and support the use of this type of formulation for the further development of efficient cancer vaccines.
Collapse
Affiliation(s)
- Danila Valmori
- *Ludwig Institute Clinical Trial Center, Columbia University, New York, NY 10032
| | | | - Valeria Tosello
- *Ludwig Institute Clinical Trial Center, Columbia University, New York, NY 10032
| | - Nina Bhardwaj
- New York University School of Medicine, New York, NY 10016
| | - Sylvia Adams
- New York University School of Medicine, New York, NY 10016
| | - David O'Neill
- New York University School of Medicine, New York, NY 10016
| | - Anna Pavlick
- New York University School of Medicine, New York, NY 10016
| | | | | | | | | | - Gregory Mears
- Division of Medical Oncology, Columbia University Medical Center, New York, NY 10032; and
| | - Susan M. Vogel
- Division of Medical Oncology, Columbia University Medical Center, New York, NY 10032; and
| | - Linda Pan
- Ludwig Institute for Cancer Research, New York, NY 10158
| | | | | | - Ralph Venhaus
- Ludwig Institute for Cancer Research, New York, NY 10158
| | - Gerd Ritter
- Ludwig Institute for Cancer Research, New York, NY 10158
| | - Lloyd J. Old
- Ludwig Institute for Cancer Research, New York, NY 10158
| | - Maha Ayyoub
- *Ludwig Institute Clinical Trial Center, Columbia University, New York, NY 10032
| |
Collapse
|