101
|
Khedri M, Rafatpanah H, Abnous K, Ramezani P, Ramezani M. Cancer immunotherapy via nucleic acid aptamers. Int Immunopharmacol 2015; 29:926-936. [PMID: 26603636 DOI: 10.1016/j.intimp.2015.10.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Revised: 10/09/2015] [Accepted: 10/13/2015] [Indexed: 02/07/2023]
Abstract
Over the past decade, immune therapy has become a standard treatment for a variety of cancers. Monoclonal antibodies, immune adjuvants and vaccines against oncogenic viruses are now well-established cancer therapies. Immune modulation is a principal element of supportive care for many high-dose chemotherapy regimens. Aptamers are short nucleic acids that bind to defined targets with high affinity and specificity. The first aptamers have been selected around two decades ago by an in vitro process named SELEX (systematic evolution of ligands by exponential enrichment). Since then, numerous aptamers with specificities for a variety of targets from small molecules to proteins or even whole cells have been selected. Targeting immunomodulatory ligands in the progressive tumor lesions of the patients would be prophylactic or therapeutic and may reduce drug-associated toxicities. A new class of inhibitory and agonistic ligands composed of short oligonucleotide (ODN) aptamers was developed recently that exhibited bioactivities comparable or superior to that of antibodies. This paper addressed progress in cancer immunotherapy with nucleic acid aptamers and highlighted recent developments either in immune system targeting or in immunotherapy methods involved aptamers. We discussed aptamer limitations when used as therapeutic agents for cancer treatment and suggested ways to overcome those limitations.
Collapse
Affiliation(s)
- Mostafa Khedri
- Department of Immunology, Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Houshang Rafatpanah
- Department of Immunology, Immunology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Pouria Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
102
|
Ipilimumab-associated Hepatitis: Clinicopathologic Characterization in a Series of 11 Cases. Am J Surg Pathol 2015; 39:1075-84. [PMID: 26034866 DOI: 10.1097/pas.0000000000000453] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Ipilimumab is a monoclonal antibody that inhibits the CTLA4 receptor on cytotoxic T lymphocytes, resulting in immune-mediated tumor cell death. Ipilimumab is most often used in the treatment of metastatic melanoma, and rarely liver toxicity necessitating cessation of treatment occurs. The aim of this study was to characterize the histologic features and clinical course of ipilimumab-associated hepatitis. Eleven patients with clinical suspicion of ipilimumab-induced hepatitis, due to the development of abnormal liver function tests (LFTs) while receiving treatment, and who underwent liver biopsy, were identified over a 6-year period. Ten patients were male and 1 female (median age 58 y), and all received 1 to 4 doses of ipilimumab. None had known preexisting liver disease. Two patients were obese, and another had a history of alcohol abuse. Viral and autoimmune serologies were negative in all patients except 1 who had a mildly elevated ANA titer. Nine biopsies showed active hepatitis with 2 distinct histologic patterns: panlobular hepatitis in 6 cases and zone 3 hepatitis in 3. The inflammatory infiltrate was similar in composition in both patterns, composed predominantly of CD8+ T lymphocytes, admixed histiocytes, scattered plasma cells, and eosinophils. Prominent histiocytic sinusoidal infiltrates were present in 7 cases and frequently formed loose histiocytic aggregates. Central vein endothelialitis was present in 8 cases. Patients in this group tended to have markedly elevated ALT, AST, and total bilirubin. Two cases did not fit into the above 2 histologic groups: 1 showed portal inflammation with cholangitis, and the other showed morphologic features indistinguishable from nonalcoholic steatohepatitis. Discontinuation of ipilimumab and administration of immunosuppressives resulted in resolution or marked improvement of LFTs in all patients within 3 months of presentation. Ipilimumab may potentially unmask previously subclinical liver disease, for example, fatty liver disease, and the diagnosis of ipilimumab-induced liver injury may only be recognized with certainty after cessation of the drug leads to normalization of LFTs. Overall, ipilimumab-associated hepatitis most often presents with a panlobular active hepatitis that resembles autoimmune hepatitis. Prominent sinusoidal histiocytic infiltrates and central vein damage with endothelialitis may be helpful histologic clues to the diagnosis of ipilimumab-associated hepatitis.
Collapse
|
103
|
Reese Z, Straubhar A, Pal SK, Agarwal N. Ipilimumab in the treatment of prostate cancer. Future Oncol 2015; 11:27-37. [PMID: 25572782 DOI: 10.2217/fon.14.196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Ipilimumab (Yervoy(®); Bristol-Myers Squibb, NY, USA) is a fully human monoclonal antibody targeting CTLA-4 and is approved for the treatment of metastatic melanoma. Preclinical and clinical studies have shown its activity in a number of different cancer types, including prostate cancer. Recently, the results from a Phase III study of ipilimumab in prostate cancer patients with prior docetaxel therapy were reported. Although the study did not meet the primary end point of improved overall survival, prespecified subset analyses suggested that ipilimumab may be more active in men with lower disease burden, which suggests that immunotherapy should be tested early in men with castration-refractory prostate cancer. Immune-related adverse events are common and most can be well managed with standard immunosuppressive algorithms.
Collapse
Affiliation(s)
- Zachary Reese
- Division of Medical Oncology, Department of Medicine, University of Utah Huntsman Cancer Institute, 2000 Circle of Hope, Ste 2123, Salt Lake City, UT 84112, USA
| | | | | | | |
Collapse
|
104
|
Buchbinder E, Hodi FS. Cytotoxic T lymphocyte antigen-4 and immune checkpoint blockade. J Clin Invest 2015; 125:3377-83. [PMID: 26325034 DOI: 10.1172/jci80012] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The relationship between cancer and the immune system is complex and provides unique therapeutic opportunities. Cytotoxic T lymphocyte antigen-4 (CTLA-4) is a regulatory molecule that suppresses T cell effector function following initial activation by costimulatory signals. Fully human monoclonal antibodies targeting CTLA-4 have been shown to increase T cell function and antitumor responses in patients with advanced metastatic melanoma. Responses observed with such immune checkpoint therapy can follow a different pattern from that seen with cytotoxic chemotherapy or targeted therapy and may continue after therapy is discontinued. In addition, the toxicities that are associated with anti-CTLA-4 therapy may differ from those of conventional therapies and consist of inflammatory events in parts of the body that do not contain cancerous cells. Early recognition of these inflammatory events and intervention is important, and the identification of predictive biomarkers continues to be an unfulfilled need in the field of immunotherapy. Combinatorial approaches with targeted therapies, radiation therapy, chemotherapy, or other immune checkpoint agonists/antagonists have the potential to increase the efficacy of CTLA-4 blockade.
Collapse
|
105
|
Boccellino M, Alaia C, Misso G, Cossu AM, Facchini G, Piscitelli R, Quagliuolo L, Caraglia M. Gene interference strategies as a new tool for the treatment of prostate cancer. Endocrine 2015; 49:588-605. [PMID: 26049369 DOI: 10.1007/s12020-015-0629-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/11/2015] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is one of the most common cancer in men. It affects older men and the incidence increases with age; the median age at diagnosis is 67 years. The diagnosis of PCa is essentially based on three tools: digital rectal exam, serum concentration of prostate specific antigen, and transrectal ultrasound-guided biopsy. Currently, the therapeutic treatments of this cancer are different and range from the prostatectomy to hormonal therapy, to radiation therapy, to immunotherapy, and to chemotherapy. However, additional efforts are required in order to find new weapons for the treatment of metastatic setting of disease. The purpose of this review is to highlight new therapeutic strategies based on gene interference; in fact, numerous siRNA and miRNA in the therapeutic treatment of PCa are reported below.
Collapse
Affiliation(s)
- Mariarosaria Boccellino
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Via L. De Crecchio, 7, 80138, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
106
|
Gupta A, De Felice KM, Loftus EV, Khanna S. Systematic review: colitis associated with anti-CTLA-4 therapy. Aliment Pharmacol Ther 2015; 42:406-17. [PMID: 26079306 DOI: 10.1111/apt.13281] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 03/11/2015] [Accepted: 05/27/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) has an important role in T-cell regulation, proliferation and tolerance. Anti-CTLA-4 agents, such as ipilimumab and tremelimumab, have been shown to prolong overall survival in patients with metastatic melanoma, and their use is being investigated in the treatment of other malignancies. Their novel immunostimulatory mechanism, however, predisposes patients to immune-related adverse effects, of which gastrointestinal effects such as diarrhoea and colitis are the most common. AIMS To discuss the existing literature and summarise the epidemiology, pathogenesis and clinical features of anti-CTLA-4-associated colitis, and to present a management algorithm for it. METHODS We searched PubMed for studies published through October 2014 using the terms 'anti-CTLA,' 'ipilimumab,' 'tremelimumab,' 'colitis,' 'gastrointestinal,' 'immune-related adverse effect,' 'immunotherapy,' 'melanoma,' and 'diarrhoea.' RESULTS Watery diarrhoea is commonly associated with anti-CTLA-4 therapy (27-54%), and symptoms occur within a few days to weeks of therapy. Diffuse acute and chronic colitis are the most common findings on endoscopy (8-22%). Concomitant infectious causes of diarrhoea must be evaluated. Most cases may be successfully managed with discontinuation of anti-CTLA-4 and conservative therapy. Those with persistent grade 2 and grade 3/4 diarrhoea should undergo endoscopic evaluation and require corticosteroid therapy. Corticosteroid-resistant cases may respond to anti-tumour necrosis factor-alpha therapy such as infliximab. Surgery is reserved for patients with bowel perforation or failure of medical therapy. CONCLUSION Given the increasing use of anti-CTLA-4 therapy, clinicians must be aware of related adverse events and their management.
Collapse
Affiliation(s)
- A Gupta
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX, USA
| | - K M De Felice
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - E V Loftus
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - S Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
107
|
van Dodewaard-de Jong JM, Verheul HM, Bloemendal HJ, de Klerk JM, Carducci MA, van den Eertwegh AJ. New Treatment Options for Patients With Metastatic Prostate Cancer: What Is The Optimal Sequence? Clin Genitourin Cancer 2015; 13:271-279. [DOI: 10.1016/j.clgc.2015.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/04/2015] [Accepted: 01/16/2015] [Indexed: 10/24/2022]
|
108
|
Callahan MK, Wolchok JD. Clinical Activity, Toxicity, Biomarkers, and Future Development of CTLA-4 Checkpoint Antagonists. Semin Oncol 2015; 42:573-86. [DOI: 10.1053/j.seminoncol.2015.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
109
|
Abstract
Tumor is one of the most common lethal diseases in the world. Current progress of therapy remains insufficient survival benefit. Tumor immunotherapies have been proposed for more than a century. With the improvement in the understanding of the role of the immune system in the tumorigenesis and immune response to tumor, immunotherapy has obtained a rapid development and plays the significant role in tumor therapy nowadays. This review designs to provide a general overview of immunotherapy in tumors. We will introduce the landmark events in the past research of immunotherapy and elaborate a range of strategies using different immune response mechanism, which have been demonstrated successfully and even some of them have been approved by US Food and Drug Administration (FDA) to certain tumor therapy. Finally, we will discuss the future direction of immunotherapy so that we can predict the possible and valuable strategies for future tumor therapy.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
110
|
Ito A, Kondo S, Tada K, Kitano S. Clinical Development of Immune Checkpoint Inhibitors. BIOMED RESEARCH INTERNATIONAL 2015; 2015:605478. [PMID: 26161407 PMCID: PMC4486755 DOI: 10.1155/2015/605478] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/07/2014] [Indexed: 01/21/2023]
Abstract
Recent progress in cancer immunotherapy has been remarkable. Most striking are the clinical development and approval of immunomodulators, also known as immune checkpoint inhibitors. These monoclonal antibodies (mAb) are directed to immune checkpoint molecules, which are expressed on immune cells and mediate signals to attenuate excessive immune reactions. Although mAbs targeting tumor associated antigens, such as anti-CD20 mAb and anti-Her2 mAb, directly recognize tumor cells and induce cell death, immune checkpoint inhibitors restore and augment the antitumor immune activities of cytotoxic T cells by blocking immune checkpoint molecules on T cells or their ligands on antigen presenting and tumor cells. Based on preclinical data, many clinical trials have demonstrated the acceptable safety profiles and efficacies of immune checkpoint inhibitors in a variety of cancers. The first in class approved immune checkpoint inhibitor is ipilimumab, an anti-CTLA-4 (cytotoxic T lymphocyte antigen-4) mAb. Two pivotal phase III randomized controlled trials demonstrated a survival benefit in patients with metastatic melanoma. In 2011, the US Food and Drug Administration (FDA) approved ipilimumab for metastatic melanoma. Several clinical trials have since investigated new agents, alone and in combination, for various cancers. In this review, we discuss the current development status of and future challenges in utilizing immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ayumu Ito
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Kohei Tada
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center (EPOC), National Cancer Center, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
111
|
Barach YS, Lee JS, Zang X. T cell coinhibition in prostate cancer: new immune evasion pathways and emerging therapeutics. Trends Mol Med 2015; 17:47-55. [PMID: 20971039 DOI: 10.1016/j.molmed.2010.09.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 09/23/2010] [Accepted: 09/23/2010] [Indexed: 12/31/2022]
Abstract
T cell-mediated adaptive immune response is controlled by both positive costimulation and negative coinhibition, generated mainly by the interaction between the B7 family and their receptor CD28 family. Coinhibition is exploited by prostate cancer as an immune evasion pathway. Overexpression of coinhibitory B7x and B7-H3 in prostate cancer correlates with poor disease outcome, whereas tumor-infiltrating immune cells have enhanced expression of PD-L1 and its receptor PD-1. New insights into the complex mechanisms governing B7 expression in the tumor microenvironment have been reported and therapies aimed at overcoming T cell coinhibition with antagonistic monoclonal antibodies are emerging as effective tumor immunotherapies. Therapies that block B7x and B7-H3, either as monotherapies or in synergism with traditional therapies, should be pursued.
Collapse
Affiliation(s)
- Yael S Barach
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jun Sik Lee
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
112
|
Current and Emerging Immunotherapies for Castration-resistant Prostate Cancer. Urology 2015; 85:976-986. [DOI: 10.1016/j.urology.2014.12.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 11/17/2022]
|
113
|
Whole Tumor Antigen Vaccines: Where Are We? Vaccines (Basel) 2015; 3:344-72. [PMID: 26343191 PMCID: PMC4494356 DOI: 10.3390/vaccines3020344] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 04/13/2015] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
With its vast amount of uncharacterized and characterized T cell epitopes available for activating CD4+ T helper and CD8+ cytotoxic lymphocytes simultaneously, whole tumor antigen represents an attractive alternative source of antigens as compared to tumor-derived peptides and full-length recombinant tumor proteins for dendritic cell (DC)-based immunotherapy. Unlike defined tumor-derived peptides and proteins, whole tumor lysate therapy is applicable to all patients regardless of their HLA type. DCs are essentially the master regulators of immune response, and are the most potent antigen-presenting cell population for priming and activating naïve T cells to target tumors. Because of these unique properties, numerous DC-based immunotherapies have been initiated in the clinics. In this review, we describe the different types of whole tumor antigens that we could use to pulse DCs ex vivo and in vivo. We also discuss the different routes of delivering whole tumor antigens to DCs in vivo and activating them with toll-like receptor agonists.
Collapse
|
114
|
MOTOSHIMA TAKANOBU, KOMOHARA YOSHIHIRO, HORLAD HASITA, TAKEUCHI ARIO, MAEDA YOSHIHIRO, TANOUE KENICHIRO, KAWANO YOSHIAKI, HARADA MAMORU, TAKEYA MOTOHIRO, ETO MASATOSHI. Sorafenib enhances the antitumor effects of anti-CTLA-4 antibody in a murine cancer model by inhibiting myeloid-derived suppressor cells. Oncol Rep 2015; 33:2947-53. [DOI: 10.3892/or.2015.3893] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/05/2015] [Indexed: 11/05/2022] Open
|
115
|
Schweizer MT, Drake CG. Immunotherapy for prostate cancer: recent developments and future challenges. Cancer Metastasis Rev 2015; 33:641-55. [PMID: 24477411 DOI: 10.1007/s10555-013-9479-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since the approval of sipuleucel-T for men with metastatic castrate resistant prostate cancer in 2010, great strides in the development of anti-cancer immunotherapies have been made. Current drug development in this area has focused primarily on antigen-specific (i.e. cancer vaccines and antibody based therapies) or checkpoint inhibitor therapies, with the checkpoint inhibitors perhaps gaining the most attention as of late. Indeed, drugs blocking the inhibitory signal generated by the engagement of cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1) found on T-cells has emerged as potent means to combat the immunosuppressive milieu. The anti-CTLA-4 monoclonal antibody ipilimumab has already been approved in advanced melanoma and two phase III trials evaluating ipilimumab in men with metastatic castrate-resistant prostate cancer are underway. A phase III trial evaluating ProstVac-VF, a poxvirus-based therapeutic prostate cancer vaccine, is also underway. While there has been reason for encouragement over the past few years, many questions regarding the use of immunotherapies remain. Namely, it is unclear what stage of disease is most likely to benefit from these approaches, how best to incorporate said treatments with each other and into our current treatment regimens and which therapy is most appropriate for which disease. Herein we review some of the recent advances in immunotherapy as related to the treatment of prostate cancer and outline some of the challenges that lie ahead.
Collapse
Affiliation(s)
- Michael T Schweizer
- Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA,
| | | |
Collapse
|
116
|
Carosella ED, Ploussard G, LeMaoult J, Desgrandchamps F. A Systematic Review of Immunotherapy in Urologic Cancer: Evolving Roles for Targeting of CTLA-4, PD-1/PD-L1, and HLA-G. Eur Urol 2015; 68:267-79. [PMID: 25824720 DOI: 10.1016/j.eururo.2015.02.032] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 02/25/2015] [Indexed: 02/06/2023]
Abstract
CONTEXT Overexpression of immune checkpoint molecules affects tumor-specific T-cell immunity in the cancer microenvironment, and can reshape tumor progression and metastasis. Antibodies targeting checkpoints could restore antitumor immunity by blocking the inhibitory receptor-ligand interaction. OBJECTIVE To analyze data and current trends in immune checkpoint targeting therapy for urologic cancers. EVIDENCE ACQUISITION Systematic literature search for clinical trials in the PubMed and Cochrane databases up to August 2014 according to Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. Endpoints included oncologic results, tumor response rates, safety, and tolerability. EVIDENCE SYNTHESIS Anti-CTLA-4 monotherapy has demonstrated biochemical responses in prostate cancer. One phase 3 trial assessing ipilimumab efficacy in castration-resistant disease was negative overall. Nevertheless, ipilimumab may significantly improve overall survival compared with placebo in subgroups of patients with favorable prognostic features. In renal cancer, phase 1 trials showed interesting stabilization or long-lasting objective response rates approaching 50% using anti-PD-1/PD-L1 drugs in heavily pretreated metastatic patients. In bladder cancer, one phase 2 trial indicated a good safety profile for ipilimumab as a neoadjuvant drug before radical cystectomy. Overall, immune-related effects such as colitis and dermatitis were common and well tolerated. CONCLUSIONS Our systematic review shows that antibodies blocking immune checkpoints offer interesting and long-lasting response rates in heavily pretreated patients with advanced urologic cancers. More promising results are currently provided by anti-CTLA-4 antibodies in prostate cancer and by PD-1/PD-L1 inhibitors in renal cancer. These should encourage new clinical trials of immune therapy combinations and immunotherapy monotherapy combined with conventional anticancer drugs. In bladder cancer, the use of targeted immunotherapy still remains underevaluated; however, preliminary results reported at recent conferences seem encouraging. PATIENT SUMMARY Data from studies support the activity and safety of immune checkpoint inhibitors in urologic cancers, alone or in combination with conventional cancer therapies. Encouraging data in other oncologic fields could translate into interesting responses in urological cancers.
Collapse
Affiliation(s)
- Edgardo D Carosella
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, Paris, France; University Paris Diderot, Sorbonne Paris Cité, UMR E_5 Institut Universitaire d'Hematologie, Saint-Louis Hospital, Paris, France.
| | | | - Joel LeMaoult
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, Paris, France; University Paris Diderot, Sorbonne Paris Cité, UMR E_5 Institut Universitaire d'Hematologie, Saint-Louis Hospital, Paris, France
| | - Francois Desgrandchamps
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, Paris, France; University Paris Diderot, Sorbonne Paris Cité, UMR E_5 Institut Universitaire d'Hematologie, Saint-Louis Hospital, Paris, France; Urology Department, Saint-Louis Hospital, Paris, France
| |
Collapse
|
117
|
Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: Enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci Transl Med 2015; 7:280sr1. [DOI: 10.1126/scitranslmed.3010274] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
118
|
Cytotoxic T-lymphocyte antigen-4 blockade in melanoma. Clin Ther 2015; 37:755-63. [PMID: 25746738 DOI: 10.1016/j.clinthera.2015.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 02/05/2023]
Abstract
PURPOSE Melanoma is an aggressive malignancy that has a complex relationship with the host immune system. Immunotherapies have long been a mainstay of melanoma therapy, and advanced therapies continue to be effective in treating this disease. Immune checkpoint blockade has proven to be a novel target in melanoma, with the approval of cytotoxic T-lymphocyte antigen-4 (CTLA-4)-targeted therapy. This review evaluates the role of CTLA-4-targeted therapies in the treatment of metastatic melanoma, with a focus on mechanisms, efficacy, toxicity, and future directions of this therapy. METHODS A search was performed in PubMed to identify relevant clinical studies that explored the clinical experience with CTLA-4-targeted therapy in melanoma. FINDINGS Signaling through CTLA-4 causes deactivation of T cells after the initial stimulatory signals. Therapies that block CTLA-4 lead to increased T-cell function and an antitumor response in patients with metastatic melanoma. The adverse event profile of these agents is different from that seen with more traditional cancer therapies and consists of dermatitis, colitis, and other autoimmune toxicities. In addition, the pattern of response is different from that seen with traditional cytotoxic therapies, with some patients experiencing initial progression followed by response and some patients having long-term durable responses. IMPLICATIONS Extensive clinical evidence supports the use of CTLA-4-targeted agents in the treatment of metastatic melanoma. The durability of response seen in patients receiving these agents has changed the landscape for patients with melanoma. Combination therapies and other agents with similar mechanisms warrant further exploration for the treatment of metastatic melanoma.
Collapse
|
119
|
Bracarda S, Altavilla A, Hamzaj A, Sisani M, Marrocolo F, Del Buono S, Danielli R. Immunologic checkpoints blockade in renal cell, prostate, and urothelial malignancies. Semin Oncol 2015; 42:495-505. [PMID: 25965369 DOI: 10.1053/j.seminoncol.2015.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Genitourinary (GU) tumors, and in particular renal cell and prostate cancer, represent one of the most dynamic areas in oncology from the scientific point of view. One of the most recent treatment approaches for GU tumors has focused on a series of molecules known as immune checkpoints and the possibility of manipulating immune responses against tumor cells by blocking these molecules with monoclonal antibodies (mAbs). Cytotoxic T lymphocyte antigen-4 (CTLA-4), and the immune checkpoint inhibitor mAbs ipilimumab and tremelimumab, represent the prototypes of this new growing class of agents called immunomodulating antibodies, while programmed death/ligand 1 (PD-1/PD-L1) also has garnered a significant interest as a new immune checkpoints to target in urothelial cancer, with the anti-PD-1/PD-L1 inhibitor mAbs nivolumab, MPDL-3280, and BMS-936559 as the first agents tested. Here we report the encouraging initial data observed in GU cancers with this new class of agents, which have reinforced the interest of investigating the therapeutic potential of the immune checkpoint modulators in large controlled trials.
Collapse
Affiliation(s)
- Sergio Bracarda
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy.
| | - Amelia Altavilla
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Alketa Hamzaj
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Michele Sisani
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Francesca Marrocolo
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Sabrina Del Buono
- Medical Oncology Unit, Department of Oncology, Ospedale San Donato, Istituto Toscano Tumori (ITT), Arezzo, Italy
| | - Riccardo Danielli
- Medical Oncology and Immunotherapy Unit, Azienda Ospedaliera Senese, University of Siena, Istituto Toscano Tumori (ITT), Siena, Italy
| |
Collapse
|
120
|
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: 16] [Impact Index Per Article: 1.8] [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
|
121
|
Finkelstein SE, Salenius S, Mantz CA, Shore ND, Fernandez EB, Shulman J, Myslicki FA, Agassi AM, Rotterman Y, DeVries T, Sims R. Combining Immunotherapy and Radiation for Prostate Cancer. Clin Genitourin Cancer 2015; 13:1-9. [DOI: 10.1016/j.clgc.2014.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/29/2014] [Accepted: 09/17/2014] [Indexed: 02/06/2023]
|
122
|
Cha E, Klinger M, Hou Y, Cummings C, Ribas A, Faham M, Fong L. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci Transl Med 2015; 6:238ra70. [PMID: 24871131 DOI: 10.1126/scitranslmed.3008211] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) blockade can promote antitumor T cell immunity and clinical responses. The mechanism by which anti-CTLA-4 antibodies induces antitumor responses is controversial. To determine the effects of CTLA-4 blockade on the T cell repertoire, we used next-generation deep sequencing to measure the frequency of individual rearranged T cell receptor β (TCRβ) genes, thereby characterizing the diversity of rearrangements, known as T cell clonotypes. CTLA-4 blockade in patients with metastatic castration-resistant prostate cancer and metastatic melanoma resulted in both expansion and loss of T cell clonotypes, consistent with a global turnover of the T cell repertoire. Overall, this treatment increased TCR diversity as reflected in the number of unique TCR clonotypes. The repertoire of clonotypes continued to evolve over subsequent months of treatment. Whereas the number of clonotypes that increased with treatment was not associated with clinical outcome, improved overall survival was associated with maintenance of high-frequency clones at baseline. In contrast, the highest-frequency clonotypes fell with treatment in patients with short overall survival. Stably maintained clonotypes included T cells having high-avidity TCR such as virus-reactive T cells. Together, these results suggest that CTLA-4 blockade induces T cell repertoire evolution and diversification. Moreover, improved clinical outcomes are associated with less clonotype loss, consistent with the maintenance of high-frequency TCR clonotypes during treatment. These clones may represent the presence of preexisting high-avidity T cells that may be relevant in the antitumor response.
Collapse
Affiliation(s)
- Edward Cha
- University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Yafei Hou
- University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Antoni Ribas
- University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Malek Faham
- Sequenta, South San Francisco, CA 94080, USA
| | - Lawrence Fong
- University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
123
|
Immunotherapy for castration-resistant prostate cancer: Progress and new paradigms. Urol Oncol 2015; 33:245-60. [PMID: 25575714 DOI: 10.1016/j.urolonc.2014.10.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/10/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND The approval of sipuleucel-T in conjunction with data from other immunotherapeutic trials for prostate cancer and other solid tumors demonstrates the potential of harnessing the patients' immune system for long-term survival. Thus, a range of therapeutic approaches are under evaluation. This review describes the rationale for immunotherapy for prostate cancer, summarizes the approaches under evaluation, and discusses sequencing options for immunotherapy in the current treatment paradigm. DESIGN References for this review were identified through searches of PubMed with the search terms "prostate cancer," "immune system," "vaccine," "immunotherapy," and "T cells." Articles were also identified through searches of the authors' own files. The final reference list was generated based on originality and relevance. RESULTS The immune system can recognize and respond to prostate tumor antigens, effected through tumor-associated antigens and tumor infiltration of immune effector cells. However, evidence also suggests that prostate tumors are adept at escaping immunological recognition, thus hypothesizing multiple therapeutic strategies. Therapeutic approaches could include vaccination and modulation of T-cell function via the blockade of checkpoint receptors such as cytotoxic T-lymphocyte antigen-4 and programmed death 1. In phase III trials, sipuleucel-T improved overall survival for an M1 patient population with castration-resistant prostate cancer and ipilimumab also did so when given after radiotherapy in a subset of better risk patients. In randomized phase II trials, prostate-specific antigen-TRICOM improved overall survival and tasquinimod improved progression-free survival. CONCLUSION Although immunotherapy has the potential to affect advanced prostate cancer, additional research is needed to (1) identify predictive or surrogate markers of activity, (2) understand which agents are clinically effective alone or in combination with other therapies, and (3) define the optimal timing for an immunotherapy to achieve maximal benefit.
Collapse
|
124
|
Burotto M, Singh N, Heery CR, Gulley JL, Madan RA. Exploiting synergy: immune-based combinations in the treatment of prostate cancer. Front Oncol 2014; 4:351. [PMID: 25566495 PMCID: PMC4264488 DOI: 10.3389/fonc.2014.00351] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 11/24/2014] [Indexed: 01/12/2023] Open
Abstract
Cancer treatment is being revolutionized by the emergence of immunotherapies such as immune checkpoint inhibitors and therapeutic cancer vaccines. Prostate cancer is amenable to such therapeutic approaches. The improved understanding of the relationship between the immune system and tumors has allowed therapeutic targeting of immune checkpoints and tumor associated antigens to be developed. Furthermore, interventions used in prostate cancer are capable of impacting the immune system. As demonstrated by preclinical data and emerging clinical data, radiation therapy, anti-androgen therapy, and chemotherapy can be used with immunotherapies to obtain synergistic results. Current and future clinical trials will further investigate these principles as immunotherapeutics are combined with each other and standard therapies for optimal clinical utility.
Collapse
Affiliation(s)
- Mauricio Burotto
- Genitourinary Malignancies Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Nishith Singh
- Indiana University Health Arnett, Lafayette, IN, USA
| | - Christopher R. Heery
- Genitourinary Malignancies Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| | - Ravi A. Madan
- Genitourinary Malignancies Branch, National Cancer Institute, National Institute of Health, Bethesda, MD, USA
| |
Collapse
|
125
|
Berman D, Korman A, Peck R, Feltquate D, Lonberg N, Canetta R. The development of immunomodulatory monoclonal antibodies as a new therapeutic modality for cancer: the Bristol-Myers Squibb experience. Pharmacol Ther 2014; 148:132-53. [PMID: 25476108 DOI: 10.1016/j.pharmthera.2014.11.017] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 11/24/2014] [Indexed: 12/19/2022]
Abstract
The discovery and increased understanding of the complex interactions regulating the immune system have contributed to the pharmacologic activation of antitumor immunity. The activity of effector cells, such as T and NK cells, is regulated by an array of activating and attenuating receptors and ligands. Agents that target these molecules can modulate immune responses by exerting antagonistic or agonistic effects. Several T- or NK-cell modulators have entered clinical trials, and two have been approved for use. Ipilimumab (Yervoy®, Bristol-Myers Squibb) and nivolumab (OPDIVO, Ono Pharmaceutical Co., Ltd./Bristol-Myers Squibb) were approved for the treatment of metastatic melanoma, in March 2011 in the United States, and in July 2014 in Japan, respectively. The clinical activity of these two antibodies has not been limited to tumor types considered sensitive to immunotherapy, and promising activity has been reported in other solid and hematologic tumors. Clinical development of ipilimumab and nivolumab has presented unique challenges in terms of safety and efficacy, requiring the establishment of new evaluation criteria for adverse events and antitumor effects. Guidelines intended to help oncologists properly manage treatment in view of these non-traditional features have been implemented. The introduction of this new modality of cancer treatment, which is meant to integrate with or replace the current standards of care, requires additional efforts in terms of optimization of treatment administration, identification of biomarkers and application of new clinical trial designs. The availability of immune modulators with different mechanisms of action offers the opportunity to establish immunological combinations as new standards of care.
Collapse
Affiliation(s)
- David Berman
- Bristol-Myers Squibb, Research and Development Division, United States
| | - Alan Korman
- Bristol-Myers Squibb, Research and Development Division, United States
| | - Ronald Peck
- Bristol-Myers Squibb, Research and Development Division, United States
| | - David Feltquate
- Bristol-Myers Squibb, Research and Development Division, United States
| | - Nils Lonberg
- Bristol-Myers Squibb, Research and Development Division, United States
| | - Renzo Canetta
- Bristol-Myers Squibb, Research and Development Division, United States.
| |
Collapse
|
126
|
Dong B, Minze LJ, Xue W, Chen W. Molecular insights into the development of T cell-based immunotherapy for prostate cancer. Expert Rev Clin Immunol 2014; 10:1547-57. [DOI: 10.1586/1744666x.2014.962515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
127
|
Fong L, Carroll P, Weinberg V, Chan S, Lewis J, Corman J, Amling CL, Stephenson RA, Simko J, Sheikh NA, Sims RB, Frohlich MW, Small EJ. Activated lymphocyte recruitment into the tumor microenvironment following preoperative sipuleucel-T for localized prostate cancer. J Natl Cancer Inst 2014; 106:dju268. [PMID: 25255802 PMCID: PMC4241888 DOI: 10.1093/jnci/dju268] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Sipuleucel-T is a US Food and Drug Administration–approved immunotherapy for asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer (mCRPC). Its mechanism of action is not fully understood. This prospective trial evaluated the direct immune effects of systemically administered sipuleucel-T on prostatic cancer tissue in the preoperative setting. Methods Patients with untreated localized prostate cancer were treated on an open-label Phase II study of sipuleucel-T prior to planned radical prostatectomy (RP). Immune infiltrates in RP specimens (posttreatment) and in paired pretreatment biopsies were evaluated by immunohistochemistry (IHC). Correlations between circulating immune response and IHC were assessed using Spearman rank order. Results Of the 42 enrolled patients, 37 were evaluable. Adverse events were primarily transient, mild-to-moderate and infusion related. Patients developed T cell proliferation and interferon-γ responses detectable in the blood following treatment. Furthermore, a greater-than-three-fold increase in infiltrating CD3+, CD4+ FOXP3-, and CD8+ T cells was observed in the RP tissues compared with the pretreatment biopsy (binomial proportions: all P < .001). This level of T cell infiltration was observed at the tumor interface, and was not seen in a control group consisting of 12 concurrent patients who did not receive any neoadjuvant treatment prior to RP. The majority of infiltrating T cells were PD-1+ and Ki-67+, consistent with activated T cells. Importantly, the magnitude of the circulating immune response did not directly correlate with T cell infiltration within the prostate based upon Spearman’s rank order correlation. Conclusions This study is the first to demonstrate a local immune effect from the administration of sipuleucel-T. Neoadjuvant sipuleucel-T elicits both a systemic antigen-specific T cell response and the recruitment of activated effector T cells into the prostate tumor microenvironment.
Collapse
Affiliation(s)
- Lawrence Fong
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF).
| | - Peter Carroll
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Vivian Weinberg
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Stephen Chan
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Jera Lewis
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - John Corman
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Christopher L Amling
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Robert A Stephenson
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Jeffrey Simko
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Nadeem A Sheikh
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Robert B Sims
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Mark W Frohlich
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| | - Eric J Small
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (LF, PC, VW, SC, JL, JS, EJS); Virginia Mason Medical Center, Seattle, WA (JC); Oregon Health & Science University, Portland, OR (CLA); University of Utah School of Medicine, Salt Lake City, UT (RAS); Dendreon Corporation, Seattle, WA (NAS, RBS, MWF)
| |
Collapse
|
128
|
Abstract
After many years of disappointments, the successful development and commercialization of the first immune checkpoint inhibitor, and regulatory approval of a dendritic cell (DC)-based cancer vaccine has led to renewed interest and enthusiasm for cancer immunotherapy. Approval of ipilimumab, an antibody targeting cytotoxic T-lymphocyte-associated antigen 4 for advanced melanoma, and sipuleucel-T, an autologous DC-based vaccine for advanced prostate cancer have brought immunotherapy to the forefront of cancer therapeutics and made the goal of long-term tumor control for patients with advanced metastatic disease seem achievable. Additionally, encouraging data from early clinical trials of other immune checkpoint inhibitors targeting programmed cell death 1 and programmed cell death ligand 1 and numerous therapeutic vaccines in development have further expanded interest in the potential of cancer immunotherapy. These recent developments represent the fruits of years of preclinical and clinical research to better understand the complex mechanisms of immune regulation and the ways in which tumors exploit those mechanisms to evade and avoid the antitumor immune response.
Collapse
Affiliation(s)
- Jeffrey S Weber
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL.
| |
Collapse
|
129
|
From bench to bedside: immunotherapy for prostate cancer. BIOMED RESEARCH INTERNATIONAL 2014; 2014:981434. [PMID: 25276838 PMCID: PMC4168152 DOI: 10.1155/2014/981434] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 08/18/2014] [Indexed: 01/20/2023]
Abstract
The mainstay therapeutic strategy for metastatic castrate-resistant prostate cancer (CRPC) continues to be androgen deprivation therapy usually in combination with chemotherapy or androgen receptor targeting therapy in either sequence, or recently approved novel agents such as Radium 223. However, immunotherapy has also emerged as an option for the treatment of this disease following the approval of sipuleucel-T by the FDA in 2010. Immunotherapy is a rational approach for prostate cancer based on a body of evidence suggesting these cancers are inherently immunogenic and, most importantly, that immunological interventions can induce protective antitumour responses. Various forms of immunotherapy are currently being explored clinically, with the most common being cancer vaccines (dendritic-cell, viral, and whole tumour cell-based) and immune checkpoint inhibition. This review will discuss recent clinical developments of immune-based therapies for prostate cancer that have reached the phase III clinical trial stage. A perspective of how immunotherapy could be best employed within current treatment regimes to achieve most clinical benefits is also provided.
Collapse
|
130
|
Liu HH, Tsai YS, Lai CL, Tang CH, Lai CH, Wu HC, Hsieh JT, Yang CR. Evolving Personalized Therapy for Castration-Resistant Prostate Cancer. Biomedicine (Taipei) 2014; 4:2. [PMID: 25520915 PMCID: PMC4264971 DOI: 10.7603/s40681-014-0002-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/31/2013] [Indexed: 12/21/2022] Open
Abstract
With advances in molecular biologic and genomic technology, detailed molecular mechanisms for development of castration-resistant prostate cancer (CRPC) have surfaced. Metastatic prostate cancer (PCa) no longer represents an end stage, with many emerging therapeutic agents approved as effective in prolonging survival of patients from either pre- or post-docetaxel stage. Given tumor heterogeneity in patients, a one-size-fits-all theory for curative therapy remains questionable. With the support of evidence from continuing clinical trials, each treatment modality has gradually been found suitable for selective best-fit patients: e.g., new androgen synthesis inhibitor arbiraterone, androgen receptor signaling inhibitor enzalutamide, sipuleucel-T immunotherapy, new taxane carbazitaxel, calcium-mimetic radium-223 radiopharmaceutical agent. Moreover, several emerging immunomodulating agents and circulating tumor cell enumeration and analysis showed promise in animal or early phase clinical trials. While the era of personalized therapy for CRPC patients is still in infancy, optimal therapeutic agents and their sequencing loom not far in the future.
Collapse
Affiliation(s)
- Hsin-Ho Liu
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Urology, Department of Surgery, Taichung Tzu Chi General Hospital, Taichung, Taiwan
- Department of Bio-Industrial Mechatronics Engineering, National Taiwan University, Taipei, Taiwan
| | - Yuh-Shyan Tsai
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Urology, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Chen-Li Lai
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Urology, Medical College and Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Tainan, Taiwan
| | - Chih-Ho Lai
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- School of Medicine, China Medical University, Tainan, Taiwan
| | - Hsi-Chin Wu
- School of Medicine, China Medical University, Tainan, Taiwan
- Department of Urology, China Medical University Hospital, Tainan, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Graduate Institute of Cancer Biology, China Medical University, Tainan, Taiwan
| | - Che-Rei Yang
- School of Medicine, China Medical University, Tainan, Taiwan
- Department of Urology, China Medical University Hospital, Tainan, Taiwan
| |
Collapse
|
131
|
Liu J, Zhang S, Li H, Cheng Y. [Advances of immunotherapy in small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:474-80. [PMID: 24949688 PMCID: PMC6000108 DOI: 10.3779/j.issn.1009-3419.2014.06.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
小细胞肺癌(small cell lung cancer, SCLC)具有复杂的异质性,由于细胞起源、发病机制和驱动基因尚不明确,SCLC的诊治进展缓慢,鲜有突破,迫切需要新的治疗策略提高SCLC疗效。肿瘤免疫治疗可提高免疫系统识别和排除肿瘤细胞的能力,且对正常组织影响轻微。目前已经开展了肿瘤疫苗、过继细胞免疫治疗、细胞因子、checkpoint抑制剂等治疗SCLC的临床研究,ipilimumab是最有前景的药物。免疫治疗有望为SCLC治疗带来新的希望,未来还需要对SCLC的异质性、免疫治疗靶点不明确、免疫治疗耐受等影响免疫治疗疗效的问题开展进一步研究。
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | - Shuang Zhang
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | - Hui Li
- Hematological Oncology Laboratory, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | | |
Collapse
|
132
|
Khan ML, Halfdanarson TR, Borad MJ. Immunotherapeutic and oncolytic viral therapeutic strategies in pancreatic cancer. Future Oncol 2014; 10:1255-75. [PMID: 24947264 DOI: 10.2217/fon.13.277] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Pancreatic adenocarcinoma is an aggressive disease with dismal outcomes despite recent advances using combination chemotherapeutic regimens. The lack of an adequate immune response to malignant cells has been identified as a factor associated with tumor aggressiveness and refractoriness to systemic treatment. Preclinical and early clinical studies have identified numerous immunotherapeutic and oncolytic viral therapeutic strategies aimed towards amplifying the immune reaction to pancreatic cancer and have established encouraging results. Promising antitumor efficacy has been observed both in vitro and in vivo with many of these approaches. These novel applications have also led to improved understanding of the process of pancreatic tumor growth and invasion, knowledge of the tumor microenvironment and have pioneered further investigations of similar therapies. Here we review both immunotherapeutic and oncolytic viral therapeutic strategies in pancreatic cancer.
Collapse
Affiliation(s)
- Meaghan L Khan
- Mayo Clinic Arizona Division of Hematology & Medical Oncology, 13400 E Shea Boulevard, Scottsdale, AZ 85259, USA
| | | | | |
Collapse
|
133
|
Roddie C, Peggs KS. Emerging options for the treatment of melanoma - focus on ipilimumab. Immunotargets Ther 2014; 3:67-78. [PMID: 27482517 PMCID: PMC4918235 DOI: 10.2147/itt.s43522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Ipilimumab is a fully human immunoglobulin subclass G1 anticytotoxic-T-lymphocyte-antigen-4 monoclonal antibody. It has been approved by the US Food and Drug Administration (FDA) and the European Medicines Agency for use in advanced melanoma following clear evidence of survival benefit in randomized Phase III studies. It is also under investigation as a treatment for other solid tumors such as renal cell, lung, and prostate cancers. The purported mechanism of antitumor activity of ipilimumab is through T-cell activation, and the side effect profile reflects this. Immune-related adverse events (irAEs) affect 60% of treated patients and 15% are defined as severe. Fortunately, most irAEs are reversible with early diagnosis and correct management. FDA approval of ipilimumab is dependent on the careful execution of a risk evaluation and mitigation strategy, with the aim of increasing awareness amongst patients and clinicians of the immunological risks of treatment, and providing algorithms for management of irAEs as they develop. Ipilimumab is one of the first immunotherapies to become widely available in the setting of solid tumors, and ongoing research aims to elucidate optimal dosing, optimal scheduling, and expanded access to ipilimumab as an adjuvant or maintenance therapy where appropriate. The identification of clinical correlates or biomarkers to identify those likely to benefit from this high-cost therapy is a top priority.
Collapse
Affiliation(s)
- Claire Roddie
- UCL Cancer Institute, Department of Hematology, London, UK
| | - Karl S Peggs
- UCL Cancer Institute, Department of Hematology, London, UK
| |
Collapse
|
134
|
A combination trial of vaccine plus ipilimumab in metastatic castration-resistant prostate cancer patients: immune correlates. Cancer Immunol Immunother 2014; 63:407-18. [PMID: 24514956 DOI: 10.1007/s00262-014-1524-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/27/2014] [Indexed: 01/05/2023]
Abstract
We recently reported the clinical results of a Phase I trial combining ipilimumab with a vaccine containing transgenes for prostate-specific antigen (PSA) and for a triad of costimulatory molecules (PROSTVAC) in patients with metastatic castration-resistant prostate cancer. Thirty patients were treated with escalating ipilimumab and a fixed dose of vaccine. Of 24 chemotherapy-naïve patients, 58 % had a PSA decline. Combination therapy did not exacerbate the immune-related adverse events associated with ipilimumab. Here, we present updated survival data and an evaluation of 36 immune cell subsets pre- and post-therapy. Peripheral blood mononuclear cells were collected before therapy, at 13 days and at 70 days post-initiation of therapy, and phenotyped by flow cytometry for the subsets of T cells, regulatory T cells, natural killer cells, and myeloid-derived suppressor cells. Associations between overall survival (OS) and immune cell subsets prior to treatment, and the change in a given immune cell subset 70 days post-initiation of therapy, were evaluated. The median OS was 2.63 years (1.77-3.45). There were trends toward associations for longer OS and certain immune cell subsets before immunotherapy: lower PD-1(+)Tim-3(NEG)CD4EM (P = 0.005, adjusted P = 0.010), higher PD-1(NEG)Tim-3(+)CD8 (P = 0.002, adjusted P = 0.004), and a higher number of CTLA-4(NEG) Tregs (P = 0.005, adjusted P = 0.010). We also found that an increase in Tim-3(+) natural killer cells post- versus pre-vaccination associated with longer OS (P = 0.0074, adjusted P = 0.015). These results should be considered as hypothesis generating and should be further evaluated in larger immunotherapy trials.
Collapse
|
135
|
Banaszynski M, Kolesar JM. Vemurafenib and ipilimumab: new agents for metastatic melanoma. Am J Health Syst Pharm 2014; 70:1205-10. [PMID: 23820456 DOI: 10.2146/ajhp120260] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE The development and place in therapy of vemurafenib and ipilimumab for the treatment of metastatic melanoma are reviewed. SUMMARY Vemurafenib is an adenosine triphosphate-competitive, reversible, and highly selective BRAF kinase inhibitor targeted at BRAF-V600E and is a first-line option for patients with BRAF-mutation-positive disease. Vemurafenib has clinically significant antitumor activity in metastatic melanoma, and response rates and overall and progression-free survival rates are improved when compared with dacarbazine. Responses also occur quickly, often within days to weeks of starting treatment. Disadvantages of vemurafenib include the short duration of response; significant skin toxicities, including skin cancers and severe photosensitivity; the need for long-term daily administration; and the potential for drug interactions. Ipilimumab is a fully human monoclonal antibody that binds to the cytotoxic T-lymphocyte-associated antigen 4 to enhance and prolong T-cell responses to elicit antitumor activity. Clinical trials have demonstrated the superiority of ipilimumab in terms of overall survival when compared with an immune stimulator and placebo. While the rate of response to ipilimumab is low, responses tended to be more durable than those achieved with vemurafenib. A disadvantage of ipilimumab is that a response may require months, making ipilimumab inappropriate as monotherapy for patients with symptomatic disease. Additional disadvantages are the adverse-effect profile and the requirement of enrollment in the risk evaluation and mitigation strategy program. CONCLUSION Vemurafenib and ipilimumab are important advances in the treatment of metastatic melanoma. Benefit is typically short lived for vemurafenib and uncommon for ipilimumab. Neither agent is curative, and clinical trials remain an alternative first-line treatment option.
Collapse
Affiliation(s)
- Megan Banaszynski
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | | |
Collapse
|
136
|
Graff JN, Puri S, Bifulco CB, Fox BA, Beer TM. Sustained complete response to CTLA-4 blockade in a patient with metastatic, castration-resistant prostate cancer. Cancer Immunol Res 2014; 2:399-403. [PMID: 24795352 DOI: 10.1158/2326-6066.cir-13-0193] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
We present the case of a man with metastatic, castration-resistant prostate cancer, who had a complete prostate-specific antigen (PSA) response after 2½ doses of ipilimumab. His treatment course was complicated by diarrhea and autoimmune hepatitis, both of which resolved within 4 months. Sera and biopsy specimens were accessed, and sera from pretreatment and day 113 were analyzed. Augmented antibody responses were detected against 11 potential tumor antigens, with responses ranging from 5- to 20-fold in day 113 sera compared with baseline. Genes that were targets of a strong antibody response (arbitrarily set at 10-fold or greater increase) were analyzed by real-time PCR for expression in the tumor biopsy cDNA. Of the top 5 genes, only 3-hydroxyisobutyryl-CoA hydrolase (HIBCH) could be identified in the amplified tumor biopsy cDNA. Using an antibody to HIBCH, immunohistochemical analysis documented strong expression of the protein. Together, these data suggest that an augmented antibody response to HIBCH, an antigen that was expressed by the patient's prostate cancer, could have contributed to the clinical response. After 16 months of PSA stability, he discontinued his androgen-suppression therapy. With the return of his testosterone, his PSA increased slightly, likely originating from his intact prostate. He has been disease free for the past 6 years without any additional therapy.
Collapse
Affiliation(s)
- Julie N Graff
- Authors' Affiliations: Department of Pathology, Providence Health System, Portland, Oregon
| | | | | | | | | |
Collapse
|
137
|
Perez-Gracia JL, Labiano S, Rodriguez-Ruiz ME, Sanmamed MF, Melero I. Orchestrating immune check-point blockade for cancer immunotherapy in combinations. Curr Opin Immunol 2014; 27:89-97. [PMID: 24485523 DOI: 10.1016/j.coi.2014.01.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/08/2014] [Accepted: 01/10/2014] [Indexed: 12/30/2022]
Abstract
Inhibitory receptors on immune system cells respond to membrane-bound and soluble ligands to abort or mitigate the intensity of immune responses by raising thresholds of activation, halting proliferation, favoring apoptosis or inhibiting/deviating effector function differentiation. Such evolutionarily selected inhibitory mechanisms are termed check-points and therefore check-point inhibitors empower any ongoing anti-cancer immune response that might have been too weak or exhausted. Monoclonal antibodies (mAb) interfering with CTLA-4-CD80/86, PD-1 - PD-L1, TIM-3-GAL9 and LAG3-MHC-II belong to this category of check-point inhibitors. The anti-CTLA-4 mAb ipilimumab has been approved for metastatic melanoma. Anti-PD-1 and anti-PD-L1 mAbs have shown extremely encouraging clinical activity. The potential of combination strategies with these agents has recently been highlighted by clinical observations on CTLA-4+PD-1 combined blockade in melanoma patients.
Collapse
Affiliation(s)
| | - Sara Labiano
- CIMA and Clinica Universidad de Navarra, Pamplona, Spain
| | | | | | - Ignacio Melero
- CIMA and Clinica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
138
|
Pezaro C, Omlin A, Bianchini D, de Bono J. New Therapies in Hormone Relapsed Disease. Prostate Cancer 2014. [DOI: 10.1002/9781118347379.ch14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
139
|
Ridolfi L, Ridolfi R. Anti-CTLA-4 therapy in melanoma: role of ipilimumab (MDX-010). ACTA ACUST UNITED AC 2014. [DOI: 10.1586/edm.09.11] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
140
|
Arlen PM, Wood LV. Prostate cancer vaccines: moving therapeutic vaccination forward in the post-Provenge™ era. Expert Rev Vaccines 2014; 11:287-302. [DOI: 10.1586/erv.11.183] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
141
|
Mittendorf EA, Sharma P. Mechanisms of T-cell inhibition: implications for cancer immunotherapy. Expert Rev Vaccines 2014; 9:89-105. [DOI: 10.1586/erv.09.144] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
142
|
Schlom J, Hodge JW, Palena C, Tsang KY, Jochems C, Greiner JW, Farsaci B, Madan RA, Heery CR, Gulley JL. Therapeutic cancer vaccines. Adv Cancer Res 2014; 121:67-124. [PMID: 24889529 PMCID: PMC6324585 DOI: 10.1016/b978-0-12-800249-0.00002-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic cancer vaccines have the potential of being integrated in the therapy of numerous cancer types and stages. The wide spectrum of vaccine platforms and vaccine targets is reviewed along with the potential for development of vaccines to target cancer cell "stemness," the epithelial-to-mesenchymal transition (EMT) phenotype, and drug-resistant populations. Preclinical and recent clinical studies are now revealing how vaccines can optimally be used with other immune-based therapies such as checkpoint inhibitors, and so-called nonimmune-based therapeutics, radiation, hormonal therapy, and certain small molecule targeted therapies; it is now being revealed that many of these traditional therapies can lyse tumor cells in a manner as to further potentiate the host immune response, alter the phenotype of nonlysed tumor cells to render them more susceptible to T-cell lysis, and/or shift the balance of effector:regulatory cells in a manner to enhance vaccine efficacy. The importance of the tumor microenvironment, the appropriate patient population, and clinical trial endpoints is also discussed in the context of optimizing patient benefit from vaccine-mediated therapy.
Collapse
Affiliation(s)
- Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kwong-Yok Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John W Greiner
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ravi A Madan
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James L Gulley
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
143
|
Sargramostim (GM-CSF) and lenalidomide in castration-resistant prostate cancer (CRPC): Results from a phase I-II clinical trial. Urol Oncol 2014; 32:33.e11-7. [DOI: 10.1016/j.urolonc.2012.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/18/2012] [Accepted: 12/27/2012] [Indexed: 11/19/2022]
|
144
|
Gogas H, Polyzos A, Kirkwood J. Immunotherapy for advanced melanoma: Fulfilling the promise. Cancer Treat Rev 2013; 39:879-85. [DOI: 10.1016/j.ctrv.2013.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 04/29/2013] [Accepted: 04/30/2013] [Indexed: 12/21/2022]
|
145
|
Ceeraz S, Nowak EC, Noelle RJ. B7 family checkpoint regulators in immune regulation and disease. Trends Immunol 2013; 34:556-63. [PMID: 23954143 PMCID: PMC3821798 DOI: 10.1016/j.it.2013.07.003] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 07/19/2013] [Accepted: 07/19/2013] [Indexed: 12/14/2022]
Abstract
Fine-tuning the immune response and maintaining tolerance to self-antigens involves a complex network of co-stimulatory and co-inhibitory molecules. The recent FDA approval of ipilimumab, a monoclonal antibody blocking cytotoxic T lymphocyte antigen (CTLA)-4, demonstrates the impact of checkpoint regulators in disease. This is reinforced by ongoing clinical trials targeting not only CTLA-4, but also the programmed death (PD)-1 and B7-H4 pathways in various disease states. Recently, two new B7 family inhibitory ligands, V-domain Ig suppressor of T cell activation (VISTA) and B7-H6 were identified. Here, we review recent understanding of B7 family members and their concerted regulation of the immune response to either self or foreign pathogens. We also discuss clinical developments in targeting these pathways in different disease settings, and introduce VISTA as a putative therapeutic target.
Collapse
Affiliation(s)
- Sabrina Ceeraz
- Geisel School of Medicine at Dartmouth, Department of Microbiology and Immunology, Norris Cotton Cancer Centre, 1 Medical Center Drive, Lebanon, New Hampshire 03756, USA
| | | | | |
Collapse
|
146
|
Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med 2013; 65:185-202. [PMID: 24188664 DOI: 10.1146/annurev-med-092012-112807] [Citation(s) in RCA: 401] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ipilimumab is the prototypical immunomodulatory antibody, approved by the FDA in 2011 for advanced melanoma on the basis of survival benefit. Since that time, we have made significant strides in optimizing this therapy: we have characterized the spectrum of immune-related adverse events and learned how to mitigate them with treatment algorithms, discovered potential biomarkers of activity, and identified the potential synergy between checkpoint modulation and other therapeutic modalities. Recent phase I trials have established the efficacy and safety of next-generation checkpoint agents, including PD-1 and PD-L1 inhibitors, across multiple tumor types. Much work lies ahead in developing these next-generation checkpoint agents, testing them in combination, and determining how to integrate them into the treatment paradigms of various tumor types.
Collapse
Affiliation(s)
- David B Page
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; , , ,
| | | | | | | | | |
Collapse
|
147
|
Kyi C, Postow MA. Checkpoint blocking antibodies in cancer immunotherapy. FEBS Lett 2013; 588:368-76. [PMID: 24161671 DOI: 10.1016/j.febslet.2013.10.015] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/12/2013] [Accepted: 10/14/2013] [Indexed: 01/05/2023]
Abstract
Cancers can be recognized by the immune system, and the immune system may regulate and even eliminate tumors. The development of checkpoint blocking antibodies, such as those directed against cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death 1 receptor (PD-1), have demonstrated significant recent promise in the treatment of an expanding list of malignancies. While both CTLA-4 and PD-1 function as negative regulators, each plays a non-redundant role in modulating immune responses. CTLA-4 attenuates the early activation of naïve and memory T cells. In contrast, PD-1 is primarily involved in modulating T cell activity in peripheral tissues via interaction with its ligands, PD-L1 and PD-L2. Unfortunately, not all patients respond to these therapies, and evaluation of biomarkers associated with clinical outcomes is ongoing. This review will examine the efficacy, toxicities, and clinical development of checkpoint blocking antibodies, including agents already approved by the US Food and Drug Administration (anti-CTLA-4, ipilimumab) or in development (anti-PD-1, PD-L1). Future studies will likely uncover new promising immunologic checkpoints to target alone or in combination with other immunotherapeutic approaches, chemotherapy, radiotherapy, and small molecules.
Collapse
Affiliation(s)
- Chrisann Kyi
- Department of Medicine, New York Presbyterian Hospital Cornell, 525 E 68th St., New York, NY 10065, United States
| | - Michael A Postow
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 300 E 66th St., New York, NY 10065, United States.
| |
Collapse
|
148
|
Pezaro C, Omlin A, Lorente D, de Bono J. Management of patients with castration-resistant disease. Hematol Oncol Clin North Am 2013; 27:1243-60, ix. [PMID: 24188261 DOI: 10.1016/j.hoc.2013.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The medical management of men with castration-resistant prostate cancer (CRPC) has changed dramatically in the last decade. Men can now access several agents developed to extend survival, delay morbidity caused by complications, and preserve quality of life. Strategies to extend survival include docetaxel and cabazitaxel, the CYP-inhibitor abiraterone acetate, the second-generation androgen receptor antagonist enzalutamide, sipuleucel-T immunotherapy, and the α-emitting radionuclide (223)radium. These novel therapies have fostered interest in translational science and a deeper understanding of the underlying biology of CRPC. This article summarizes clinical data and unresolved issues in the use of current and emerging CRPC therapies.
Collapse
Affiliation(s)
- Carmel Pezaro
- Prostate Cancer Targeted Therapy Group and Drug Development Unit, The Royal Marsden NHS Foundation Trust, The Institute of Cancer Research, Downs Road, Sutton, Surrey SM2 5PT, UK
| | | | | | | |
Collapse
|
149
|
Abstract
Over the past 7 decades androgen-deprivation therapy (ADT) has been the cornerstone of treatment for metastatic non-castrate prostate cancer (NCPC); however, the mechanisms to achieve this goal have evolved over time to include not only bilateral orchiectomy and estrogens, but also gonadotropin-releasing hormone (GnRH) agonists, antagonists, and the inclusion of androgen receptor (AR) blockade. Despite treatment with ADT, most men will progress to castrate-resistant prostate cancer (CRPC). Over the last decade many new treatment options for CRPC have emerged. These new treatments also could have a meaningful role earlier in NCPC. In this review, we outline the biologic drivers of NCPC, review current standard therapy available for NCPC, and discuss the evolving role of new therapeutics in metastatic disease.
Collapse
Affiliation(s)
- Phillip L Palmbos
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, MI 48109-5946, USA
| | | |
Collapse
|
150
|
Romero PJ, Withington T, Marincola F. Immune evasion in acute myeloid leukemia: current concepts and future directions. J Immunother Cancer 2013; 1:1/1/13. [PMID: 24353898 PMCID: PMC3864190 DOI: 10.1186/2051-1426-1-13] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 05/14/2013] [Indexed: 01/22/2023] Open
Abstract
Immune responses generated against malignant cells have the potential to inhibit tumor growth, or even eliminate transformed cells before a tumor forms. However, immune tolerance mechanisms that normally protect healthy tissues from autoimmune damage pose a formidable barrier to the development of effective anti-tumor immunity. Because malignant cells are derived from self-tissues, the majority of defined tumor antigens are either shared or aberrantly expressed self-proteins. Eliciting productive T cell responses against such proteins is challenging, as most high-affinity, self-reactive T cells are purged during thymic selection. Some T cells capable of tumor antigen recognition escape thymic deletion, but are functionally inhibited by peripheral tolerance mechanisms which limit their ability to attack a developing malignancy. Alternatively, some tumors express antigens derived from mutated self-proteins, viral proteins or self proteins expressed only during embryonic development. These antigens are recognized by the immune system as foreign and could be recognized by a relatively large number of peripheral T cells. Even in this scenario, tumors evade otherwise effective T cell responses by employing potent immunosuppressive mechanisms within their local environment. In the setting for solid malignancies, such as melanoma, a growing number of putative immune evasion mechanisms have been characterized. However, acute myeloid leukemia (AML) is a systemic disease, and the pathways it exploits to subvert the host immune response may be quite different than those of a solid tumor. Much remains unknown regarding the immune escape mechanisms promoted by AML, and whether efforts to thwart tolerance may influence the progression of this disease. Here, we review current concepts of immune evasion in AML, and speculate how potentially effective immunotherapeutic strategies might be developed to reverse immune tolerance in leukemia patients in the future.
Collapse
Affiliation(s)
- Pedro J Romero
- Ludwig Center for Cancer Research, Lausanne, Switzerland
| | - Tara Withington
- Society for Immunotherapy of Cancer (SITC), Milwaukee, WI, USA
| | | |
Collapse
|