51
|
Khoja L, Day D, Wei-Wu Chen T, Siu LL, Hansen AR. Tumour- and class-specific patterns of immune-related adverse events of immune checkpoint inhibitors: a systematic review. Ann Oncol 2018; 28:2377-2385. [PMID: 28945858 DOI: 10.1093/annonc/mdx286] [Citation(s) in RCA: 576] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Immune checkpoint inhibitor (ICI) monoclonal antibodies (mAbs) targeting cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1) or its ligand (PD-L1) produce unique toxicity profiles. The objective of this review was to identify patterns and incidence of immune-related adverse events (irAE) based on tumour type and ICI class. Methods Medline, EMBASE and COCHRANE databases were searched to identify prospective monotherapy trials of ICIs from 2003 to November 2015. Paired reviewers selected studies for inclusion and extracted data. Odds ratio (OR), χ2 tests and multivariable regression models were used to analyse for effect size and associations. Results We identified 48 trials (6938 patients), including 26 CTLA-4, 17 PD-1, 2 PD-L1 trials, and 3 studies tested both CTLA-4 and PD-1. Grade 3/4 irAE were more common with CTLA-4 mAbs compared with PD-1 (31% versus 10%). All grades colitis (OR 8.7, 95% CI 5.8-12.9), hypophysitis (OR 6.5, 95% CI 3.0-14.3) and rash (OR 2.0, 95% CI 1.8-2.3) were more frequent with CTLA-4 mAbs; whereas pneumonitis (OR 6.4, 95% CI 3.2-12.7), hypothyroidism (OR 4.3, 95% CI 2.9-6.3), arthralgia (OR 3.5, 95% CI 2.6-4.8) and vitiligo (OR 3.5, 95% CI 2.3-5.3) were more common with PD-1 mAbs. Comparison of irAE from the three most studied tumour types in PD-1 mAbs trials [melanoma (n = 2048), non-small-cell lung cancer (n = 1030) and renal cell carcinoma (n = 573)] showed melanoma patients had a higher frequency of gastrointestinal and skin irAE and lower frequency of pneumonitis. Discussion CTLA-4 and PD-1 mAbs have distinct irAE profiles. Different immune microenvironments may drive histology-specific irAE patterns. Other tumour-dependent irAE profiles may be identified as data emerge from ICI trials.
Collapse
Affiliation(s)
- L Khoja
- Clinical Development Unit, Early Clinical Development, AstraZeneca UK plc, Melbourn Science Park, Melbourn, Hertfordshire;; Medical Oncology, Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, UK
| | - D Day
- Drug Development Program, Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto;; Department of Medicine, University of Toronto, Toronto;; Ontario Institute for Cancer Research (OICR), Toronto, Canada
| | - T Wei-Wu Chen
- Department of Oncology, National Taiwan University Hospital, Taipei;; National Taiwan University Cancer Center, Taipei;; Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - L L Siu
- Drug Development Program, Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto;; Department of Medicine, University of Toronto, Toronto
| | - A R Hansen
- Drug Development Program, Division of Medical Oncology and Haematology, Princess Margaret Cancer Centre, Toronto;; Department of Medicine, University of Toronto, Toronto;.
| |
Collapse
|
52
|
Hung AL, Maxwell R, Theodros D, Belcaid Z, Mathios D, Luksik AS, Kim E, Wu A, Xia Y, Garzon-Muvdi T, Jackson C, Ye X, Tyler B, Selby M, Korman A, Barnhart B, Park SM, Youn JI, Chowdhury T, Park CK, Brem H, Pardoll DM, Lim M. TIGIT and PD-1 dual checkpoint blockade enhances antitumor immunity and survival in GBM. Oncoimmunology 2018; 7:e1466769. [PMID: 30221069 PMCID: PMC6136875 DOI: 10.1080/2162402x.2018.1466769] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/12/2022] Open
Abstract
The use of inhibitory checkpoint blockade in the management of glioblastoma has been studied in both preclinical and clinical settings. TIGIT is a novel checkpoint inhibitor recently discovered to play a role in cancer immunity. In this study, we sought to determine the effect of anti-PD-1 and anti-TIGIT combination therapy on survival in a murine glioblastoma (GBM) model, and to elucidate the underlying immune mechanisms. Using mice with intracranial GL261-luc+ tumors, we found that TIGIT expression was upregulated on CD8+ and regulatory T cells (Tregs) in the brain compared to draining cervical lymph nodes (CLN) and spleen. We then demonstrated that treatment using anti-PD-1 and anti-TIGIT dual therapy significantly improved survival compared to control and monotherapy groups. The therapeutic effect was correlated with both increased effector T cell function and downregulation of suppressive Tregs and tumor-infiltrating dendritic cells (TIDCs). Clinically, TIGIT expression on tumor-infiltrating lymphocytes was shown to be elevated in patient GBM samples, suggesting that the TIGIT pathway may be a valuable therapeutic target. Expression of the TIGIT ligand, PVR, further portended a poor survival outcome in patients with low-grade glioma. We conclude that anti-TIGIT is an effective treatment strategy against murine GBM when used in combination with anti-PD-1, improving overall survival via modifications of both the T cell and myeloid compartments. Given evidence of PVR expression on human GBM cells, TIGIT presents as a promising immune therapeutic target in the management of these patients.
Collapse
Affiliation(s)
- Alice L Hung
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Russell Maxwell
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Debebe Theodros
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Zineb Belcaid
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Andrew S Luksik
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Eileen Kim
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Adela Wu
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Yuanxuan Xia
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | | | | | - Xiaobu Ye
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | | | | | | | - Su-Myeong Park
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea
| | - Je-In Youn
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Tamrin Chowdhury
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
53
|
Caponnetto S, Draghi A, Borch TH, Nuti M, Cortesi E, Svane IM, Donia M. Cancer immunotherapy in patients with brain metastases. Cancer Immunol Immunother 2018; 67:703-711. [PMID: 29520474 PMCID: PMC11028279 DOI: 10.1007/s00262-018-2146-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 03/05/2018] [Indexed: 11/26/2022]
Abstract
The exclusion of "real-world" patients from registration clinical trials of cancer immunotherapy represents a significant emerging issue. For instance, a large fraction of cancer patients develops brain metastases during the course of the disease, but results from large prospective clinical trials investigating this considerable proportion of the cancer patient population are currently lacking. To provide a useful tool for the clinician in a "real-world" setting, we have reviewed the available literature regarding the safety and efficacy of immune check-point inhibitors in patients with cancer metastatic to the brain. Overall, these data provide encouraging evidence that these therapeutic agents can induce intracranial objective responses, particularly in patients with asymptomatic and previously untreated brain metastases. Larger prospective studies are needed to confirm these initial results.
Collapse
Affiliation(s)
- Salvatore Caponnetto
- Cell Therapy Unit and Laboratory of Tumor Immunology, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
- Division of Medical Oncology B, Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Arianna Draghi
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Troels Holz Borch
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark
| | - Marianna Nuti
- Division of Medical Oncology B, Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Enrico Cortesi
- Cell Therapy Unit and Laboratory of Tumor Immunology, Department of Experimental Medicine, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Inge Marie Svane
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark.
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.
| | - Marco Donia
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730, Herlev, Denmark.
- Department of Oncology, Herlev Hospital, University of Copenhagen, Herlev, Denmark.
| |
Collapse
|
54
|
Viala M, Vinches M, Alexandre M, Mollevi C, Durigova A, Hayaoui N, Homicsko K, Cuenant A, Gongora C, Gianni L, Tosi D. Strategies for clinical development of monoclonal antibodies beyond first-in-human trials: tested doses and rationale for dose selection. Br J Cancer 2018; 118:679-697. [PMID: 29438365 PMCID: PMC5846071 DOI: 10.1038/bjc.2017.473] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 11/29/2017] [Accepted: 11/30/2017] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Our previous survey on first-in-human trials (FIHT) of monoclonal antibodies (mAbs) showed that, due to their limited toxicity, the recommended phase II dose (RP2D) was only tentatively defined. METHODS We identified, by MEDLINE search, articles on single-agent trials of mAbs with an FIHT included in our previous survey. For each mAb, we examined tested dose(s) and dose selection rationale in non-FIHTs (NFIHTs). We also assessed the correlation between doses tested in the registration trials (RTs) of all FDA-approved mAbs and the corresponding FIHT results. RESULTS In the 37 dose-escalation NFIHTs, the RP2D indication was still poorly defined. In phase II-III NFIHTs (n=103 on 37 mAbs), the FIHT RP2D was the only dose tested for five mAbs. For 16 mAbs, only doses different from the FIHT RP2D or the maximum administered dose (MAD) were tested and the dose selection rationale infrequently indicated. In the 60 RTs on 27 FDA-approved mAbs with available FIHT, the FIHT RP2D was tested only for two mAbs, and RT doses were much lower than the FIHT MAD. CONCLUSIONS The rationale beyond dose selection in phase II and III trials of mAbs is often unclear in published articles and not based on FIHT data.
Collapse
Affiliation(s)
- Marie Viala
- Institut du Cancer de Montpellier, Montpellier, France
| | - Marie Vinches
- Institut du Cancer de Montpellier, Montpellier, France
| | | | | | | | - Nadia Hayaoui
- Institut du Cancer de Montpellier, Montpellier, France
| | | | - Alice Cuenant
- Institut du Cancer de Montpellier, Montpellier, France
| | - Céline Gongora
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Montpellier, France
| | - Luca Gianni
- San Raffaele – Scientific Institute, Milan, Italy
| | - Diego Tosi
- Institut du Cancer de Montpellier, Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier, Inserm U1194, Montpellier, France
| |
Collapse
|
55
|
Roufas C, Chasiotis D, Makris A, Efstathiades C, Dimopoulos C, Zaravinos A. The Expression and Prognostic Impact of Immune Cytolytic Activity-Related Markers in Human Malignancies: A Comprehensive Meta-analysis. Front Oncol 2018. [PMID: 29515971 PMCID: PMC5826382 DOI: 10.3389/fonc.2018.00027] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Recently, immune-checkpoint blockade has shown striking clinical results in different cancer patients. However, a significant inter-individual and inter-tumor variability exists among different cancers. The expression of the toxins granzyme A (GZMA) and perforin 1 (PRF1), secreted by effector cytotoxic T cells and natural killer (NK) cells, were recently used as a denominator of the intratumoral immune cytolytic activity (CYT). These levels are significantly elevated upon CD8+ T-cell activation as well as during a productive clinical response against immune-checkpoint blockade therapies. Still, it is not completely understood how different tumors induce and adapt to immune responses. Methods Here, we calculated the CYT across different cancer types and focused on differences between primary and metastatic tumors. Using data from 10,355, primary tumor resection samples and 2,787 normal samples that we extracted from The Cancer Genome Atlas and Genotype-Tissue Expression project databases, we screened the variation of CYT across 32 different cancer types and 28 different normal tissue types. We correlated the cytolytic levels in each cancer type with the corresponding patient group's overall survival, the expression of several immune-checkpoint molecules, as well as with the load of tumor-infiltrating lymphocytes (TILs), and tumor-associated neutrophils (TANs) in these tumors. Results We found diverse levels of CYT across different cancer types, with highest levels in kidney, lung, and cervical cancers, and lowest levels in glioma, adrenocortical carcinoma (ACC), and uveal melanoma. GZMA protein was either lowly expressed or absent in at least half of these tumors; whereas PRF1 protein was not detected in almost any of the different tumor types, analyzing tissue microarrays from 20 different tumor types. CYT was significantly higher in metastatic skin melanoma and correlated significantly to the TIL load. In TCGA-ACC, skin melanoma, and bladder cancer, CYT was associated with an improved patient outcome and high levels of both GZMA and PRF1 synergistically affected patient survival in these cancers. In bladder, breast, colon, esophageal, kidney, ovarian, pancreatic, testicular, and thyroid cancers, high CYT was accompanied by upregulation of at least one immune-checkpoint molecule, indicating that similar to melanoma and prostate cancer, immune responses in cytolytic-high tumors elicit immune suppression in the tumor microenvironment. Conclusion Overall, our data highlight the existence of diverse levels of CYT across different cancer types and suggest that along with the existence of complicated associations among various tumor-infiltrated immune cells, it is capable to promote or inhibit the establishment of a permissive tumor microenvironment, depending on the cancer type. High levels of immunosuppression seem to exist in several tumor types.
Collapse
Affiliation(s)
- Constantinos Roufas
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus.,The Center for Risk and Decision Sciences (CERIDES), Department of Computer Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Dimitrios Chasiotis
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Anestis Makris
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Christodoulos Efstathiades
- The Center for Risk and Decision Sciences (CERIDES), Department of Computer Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Christos Dimopoulos
- The Center for Risk and Decision Sciences (CERIDES), Department of Computer Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, Biomedical Sciences Program, School of Sciences, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
56
|
Redelman-Sidi G, Michielin O, Cervera C, Ribi C, Aguado JM, Fernández-Ruiz M, Manuel O. ESCMID Study Group for Infections in Compromised Hosts (ESGICH) Consensus Document on the safety of targeted and biological therapies: an infectious diseases perspective (Immune checkpoint inhibitors, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors). Clin Microbiol Infect 2018; 24 Suppl 2:S95-S107. [PMID: 29427804 DOI: 10.1016/j.cmi.2018.01.030] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/18/2018] [Accepted: 01/27/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND The present review is part of the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Infections in Compromised Hosts (ESGICH) consensus document on the safety of targeted and biological therapies. AIMS To review, from an infectious diseases perspective, the safety profile of immune checkpoint inhibitors, LFA-3-targeted agents, cell adhesion inhibitors, sphingosine-1-phosphate receptor modulators and proteasome inhibitors, and to suggest preventive recommendations. SOURCES Computer-based Medline searches with MeSH terms pertaining to each agent or therapeutic family. CONTENT T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death (PD)-1/PD-1 ligand 1 (PD-L1)-targeted agents do not appear to intrinsically increase the risk of infection but can induce immune-related adverse effects requiring additional immunosuppression. Although CD4+ T-cell lymphopenia is associated with alefacept, no opportunistic infections have been observed. Progressive multifocal leukoencephalopathy (PML) may occur during therapy with natalizumab (anti-α4-integrin monoclonal antibody (mAb)) and efalizumab (anti-CD11a mAb), but no cases have been reported to date with vedolizumab (anti-α4β7 mAb). In patients at high risk for PML (positive anti-JC polyomavirus serology with serum antibody index >1.5 and duration of therapy ≥48 months), the benefit-risk ratio of continuing natalizumab should be carefully considered. Fingolimod induces profound peripheral blood lymphopenia and increases the risk of varicella zoster virus (VZV) infection. Prophylaxis with (val)acyclovir and VZV vaccination should be considered. Proteasome inhibitors also increase the risk of VZV infection, and antiviral prophylaxis with (val)acyclovir is recommended. Anti-Pneumocystis prophylaxis may be considered in myeloma multiple patients with additional risk factors (i.e. high-dose corticosteroids). IMPLICATIONS Clinicians should be aware of the risk of immune-related adverse effects and PML in patients receiving immune checkpoint and cell adhesion inhibitors respectively.
Collapse
Affiliation(s)
- G Redelman-Sidi
- Service of Infectious Disease, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA.
| | - O Michielin
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - C Cervera
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - C Ribi
- Department of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - J M Aguado
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - M Fernández-Ruiz
- Unit of Infectious Diseases, Hospital Universitario '12 de Octubre', Instituto de Investigación Hospital '12 de Octubre' (i+12), Madrid, Spain; Spanish Network for Research in Infectious Diseases (REIPI RD16/0016), Instituto de Salud Carlos III, Madrid, Spain
| | - O Manuel
- Department of Infectious Diseases, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
57
|
Bowman KM, Kumthekar P. Medical management of brain metastases and leptomeningeal disease in patients with breast carcinoma. Future Oncol 2018; 14:391-407. [DOI: 10.2217/fon-2017-0255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Breast cancer is the most common malignancy among women and accounts for the second highest number of cancer-related deaths. With patients surviving longer due to advances in systemic control, the incidence of CNS involvement is increasing; however, the management of CNS metastases has not undergone parallel advancements. The blood–brain barrier limits the efficacy of most systemic chemotherapies, and the utilization of surgery and radiation beyond first-line therapy is limited. We will explore the recent developments in the medical management of breast cancer brain metastasis. Beyond traditional chemotherapy, we will also discuss targeted therapies and immunotherapies which may provide a survival benefit to this population and thus, offer further treatment options and a path for future research and treatment advances.
Collapse
Affiliation(s)
- Kelsey M Bowman
- Department of Neurology, Northwestern University Feinberg School of Medicine, Abbott Hall, Chicago, IL 60607, USA
| | - Priya Kumthekar
- Department of Neurology, Robert H Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60607, USA
| |
Collapse
|
58
|
Cuesta-Mateos C, Alcaraz-Serna A, Somovilla-Crespo B, Muñoz-Calleja C. Monoclonal Antibody Therapies for Hematological Malignancies: Not Just Lineage-Specific Targets. Front Immunol 2018; 8:1936. [PMID: 29387053 PMCID: PMC5776327 DOI: 10.3389/fimmu.2017.01936] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/15/2017] [Indexed: 12/12/2022] Open
Abstract
Today, monoclonal antibodies (mAbs) are a widespread and necessary tool for biomedical science. In the hematological cancer field, since rituximab became the first mAb approved by the Food and Drug Administration for the treatment of B-cell malignancies, a number of effective mAbs targeting lineage-specific antigens (LSAs) have been successfully developed. Non-LSAs (NLSAs) are molecules that are not restricted to specific leukocyte subsets or tissues but play relevant pathogenic roles in blood cancers including the development, proliferation, survival, and refractoriness to therapy of tumor cells. In consequence, efforts to target NLSAs have resulted in a plethora of mAbs-marketed or in development-to achieve different goals like neutralizing oncogenic pathways, blocking tumor-related chemotactic pathways, mobilizing malignant cells from tumor microenvironment to peripheral blood, modulating immune-checkpoints, or delivering cytotoxic drugs into tumor cells. Here, we extensively review several novel mAbs directed against NLSAs undergoing clinical evaluation for treating hematological malignancies. The review focuses on the structure of these antibodies, proposed mechanisms of action, efficacy and safety profile in clinical studies, and their potential applications in the treatment of hematological malignancies.
Collapse
Affiliation(s)
- Carlos Cuesta-Mateos
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
- IMMED S.L., Immunological and Medicinal Products, Madrid, Spain
| | - Ana Alcaraz-Serna
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| | - Beatriz Somovilla-Crespo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Servicio de Inmunología, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
| |
Collapse
|
59
|
Rowshanravan B, Halliday N, Sansom DM. CTLA-4: a moving target in immunotherapy. Blood 2018; 131:58-67. [PMID: 29118008 PMCID: PMC6317697 DOI: 10.1182/blood-2017-06-741033] [Citation(s) in RCA: 719] [Impact Index Per Article: 119.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/31/2017] [Indexed: 02/08/2023] Open
Abstract
CD28 and CTLA-4 are members of a family of immunoglobulin-related receptors that are responsible for various aspects of T-cell immune regulation. The family includes CD28, CTLA-4, and ICOS as well as other proteins, including PD-1, BTLA, and TIGIT. These receptors have both stimulatory (CD28, ICOS) and inhibitory roles (CTLA-4, PD-1, BTLA, and TIGIT) in T-cell function. Increasingly, these pathways are targeted as part of immune modulatory strategies to treat cancers, referred to generically as immune checkpoint blockade, and conversely to treat autoimmunity and CTLA-4 deficiency. Here, we focus on the biology of the CD28/CTLA-4 pathway as a framework for understanding the impacts of therapeutic manipulation of this pathway.
Collapse
Affiliation(s)
- Behzad Rowshanravan
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - Neil Halliday
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| | - David M Sansom
- Institute of Immunity and Transplantation, Division of Infection & Immunity, University College London, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
60
|
Postel-Vinay S, Soria JC. Immune Therapies in Phase 1 Trials. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
61
|
Cha SW, Bonissone S, Na S, Pevzner PA, Bafna V. The Antibody Repertoire of Colorectal Cancer. Mol Cell Proteomics 2017; 16:2111-2124. [PMID: 29046389 PMCID: PMC5724175 DOI: 10.1074/mcp.ra117.000397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Indexed: 12/31/2022] Open
Abstract
Immunotherapy is becoming increasingly important in the fight against cancers, using and manipulating the body's immune response to treat tumors. Understanding the immune repertoire-the collection of immunological proteins-of treated and untreated cells is possible at the genomic, but technically difficult at the protein level. Standard protein databases do not include the highly divergent sequences of somatic rearranged immunoglobulin genes, and may lead to miss identifications in a mass spectrometry search. We introduce a novel proteogenomic approach, AbScan, to identify these highly variable antibody peptides, by developing a customized antibody database construction method using RNA-seq reads aligned to immunoglobulin (Ig) genes.AbScan starts by filtering transcript (RNA-seq) reads that match the template for Ig genes. The retained reads are used to construct a repertoire graph using the "split" de Bruijn graph: a graph structure that improves on the standard de Bruijn graph to capture the high diversity of Ig genes in a compact manner. AbScan corrects for sequencing errors, and converts the graph to a format suitable for searching with MS/MS search tools. We used AbScan to create an antibody database from 90 RNA-seq colorectal tumor samples. Next, we used proteogenomic analysis to search MS/MS spectra of matched colorectal samples from the Clinical Proteomic Tumor Analysis Consortium (CPTAC) against the AbScan generated database. AbScan identified 1,940 distinct antibody peptides. Correlating with previously identified Single Amino-Acid Variants (SAAVs) in the tumor samples, we identified 163 pairs (antibody peptide, SAAV) with significant cooccurrence pattern in the 90 samples. The presence of coexpressed antibody and mutated peptides was correlated with survival time of the individuals. Our results suggest that AbScan (https://github.com/csw407/AbScan.git) is an effective tool for a proteomic exploration of the immune response in cancers.
Collapse
Affiliation(s)
- Seong Won Cha
- From the ‡Department of Electrical and Computer Engineering, University of California, San Diego, La Jolla, California
| | | | - Seungjin Na
- ¶Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California 92037
| | - Pavel A Pevzner
- ¶Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California 92037
| | - Vineet Bafna
- ¶Department of Computer Science and Engineering, University of California, San Diego, La Jolla, California 92037
| |
Collapse
|
62
|
Amaral T, Meraz-Torres F, Garbe C. Immunotherapy in managing metastatic melanoma: which treatment when? Expert Opin Biol Ther 2017; 17:1523-1538. [PMID: 28891339 DOI: 10.1080/14712598.2017.1378640] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Ten to fifteen percent of melanoma patients develop distant or unresectable metastasis requiring systemic treatment. Around 45% of the patients diagnosed with metastatic cutaneous melanoma harbor a BRAFV600 mutation and derive benefit from combined targeted therapy with MAPK pathway inhibitors. These offer a rapid response that translates into improvement of symptoms and increased quality of life. However, resistance often develops with subsequent progressive disease. Immunotherapy with checkpoint inhibitors may be offered to BRAF-mutated and wild-type patients and is associated with longer and durable responses that can continue over years. Areas covered: In this review, the authors discuss the late evidence for targeted and immunotherapy in melanoma patients, as well as therapy sequencing. Immunotherapy in special populations is also addressed. Expert opinion: Effective treatments are currently available. However, there are still unanswered questions of the best therapy sequence, the clear superiority of combined immunotherapy versus monotherapy in all patients, and therapy duration. Since different promising treatments will become available, clinical trials comparing the diverse options in terms of safety, efficacy and cost- effectiveness are required to make the right decisions. Consequently, patients should be encouraged to participate in clinical trials, whenever possible.
Collapse
Affiliation(s)
- Teresa Amaral
- a Center for Dermatooncology, Department of Dermatology , University Hospital Tuebingen , Tuebingen , Germany
- b Portuguese Air Force Health Direction , Lisbon , Portugal
| | - Francisco Meraz-Torres
- a Center for Dermatooncology, Department of Dermatology , University Hospital Tuebingen , Tuebingen , Germany
| | - Claus Garbe
- a Center for Dermatooncology, Department of Dermatology , University Hospital Tuebingen , Tuebingen , Germany
| |
Collapse
|
63
|
Brown ZJ, Heinrich B, Steinberg SM, Yu SJ, Greten TF. Safety in treatment of hepatocellular carcinoma with immune checkpoint inhibitors as compared to melanoma and non-small cell lung cancer. J Immunother Cancer 2017; 5:93. [PMID: 29157287 PMCID: PMC5697069 DOI: 10.1186/s40425-017-0298-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 10/27/2017] [Indexed: 12/26/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a major health problem worldwide with increasing incidence rates. As HCC traditionally occurs in chronically inflamed livers, this inflammation aids to drive oncogenesis and often renders these lesions to be immunogenic and therefore potential targets for immunotherapy. As patients with HCC generally have underlying liver dysfunction, we sought to determine if immune checkpoint inhibitors were safe to use in patients with HCC as compared to melanoma and non-small cell lung cancer (NSCLC) in terms of the gastrointestinal side effects of elevation of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and diarrhea as well as patients who drop out of the study due to drug toxicity and death secondary to drug toxicity. Methods A literature review was performed for clinical trials that have been completed with single agent immune checkpoint inhibitors for patients with HCC, melanoma, and NSCLC. Gastrointestinal related adverse events including elevation of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and diarrhea were analyzed as well as those patients who were taken off therapy secondary to drug related toxicity and patients who died as a result of therapy. Results We found that although patients with HCC treated with immune checkpoint inhibitors have a substantial increase in AST/ALT as compared to patients with melanoma and NSCLC, this does not cause the patients to come off therapy or cause death secondary to drug toxicity. Conclusions We propose immune checkpoint inhibitors are safe to pursue in the treatment of HCC.
Collapse
Affiliation(s)
- Zachary J Brown
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Bernd Heinrich
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Su Jong Yu
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tim F Greten
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
64
|
Not all immune-checkpoint inhibitors are created equal: Meta-analysis and systematic review of immune-related adverse events in cancer trials. Crit Rev Oncol Hematol 2017; 119:1-12. [DOI: 10.1016/j.critrevonc.2017.09.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/13/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023] Open
|
65
|
Davoodzadeh Gholami M, Kardar GA, Saeedi Y, Heydari S, Garssen J, Falak R. Exhaustion of T lymphocytes in the tumor microenvironment: Significance and effective mechanisms. Cell Immunol 2017; 322:1-14. [PMID: 29079339 DOI: 10.1016/j.cellimm.2017.10.002] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/23/2022]
Abstract
T lymphocytes play crucial roles in adaptive immune responses to tumors. However, due to different tolerance mechanisms and inhibitory effects of the tumor microenvironment (TME) on T cells, responses to tumors are insufficient. In fact, cellular and molecular suppressive mechanisms repress T cell responses in the TME, resulting in senescent, anergic and exhausted lymphocytes. Exhaustion is a poor responsive status of T cells, with up-regulated expression of inhibitory receptors, decreased production of effective cytokines, and reduced cytotoxic activity. Low immunogenicity of tumor antigens and inadequate presentation of tumor-specific antigens results in inappropriate activation of naive T lymphocytes against tumor antigens. Moreover, when effector cytotoxic T cells enter TME, they encounter a complicated network of cells and cytokines that suppress their effectiveness and turn them into exhausted T cells. Thus, the mechanism of T cell exhaustion in cancer is different from that in chronic infections. In this review we will discuss the main components such as inhibitory receptors, inflammatory cells, stromal cells, cytokine milieu as well as environmental and metabolic conditions in TME which play role in development of exhaustion. Furthermore, recent therapeutic methods available to overcome exhaustion will be discussed.
Collapse
Affiliation(s)
- Mohammad Davoodzadeh Gholami
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yousef Saeedi
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Sahel Heydari
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
66
|
Callahan MK, Postow MA, Wolchok JD. Targeting T Cell Co-receptors for Cancer Therapy. Immunity 2017; 44:1069-78. [PMID: 27192570 DOI: 10.1016/j.immuni.2016.04.023] [Citation(s) in RCA: 368] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Indexed: 12/15/2022]
Abstract
Checkpoint-blocking antibodies can generate potent anti-tumor responses by encouraging the immune system to seek and destroy cancer cells. At this time, the United States Food and Drug Administration has approved three checkpoint-blocking antibodies in three disease indications, and additional approvals are expected to broaden the clinical scope of immunotherapy. Herein, we review the clinical development of CTLA-4-, PD-1-, and PD-L1-blocking antibodies across tumor types and briefly discuss areas of active investigation of potential biomarkers.
Collapse
Affiliation(s)
- Margaret K Callahan
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, Cornell University, New York, NY 10065, USA.
| | - Michael A Postow
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Jedd D Wolchok
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Medical College, Cornell University, New York, NY 10065, USA; Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
67
|
Pérez-De-Lis M, Retamozo S, Flores-Chávez A, Kostov B, Perez-Alvarez R, Brito-Zerón P, Ramos-Casals M. Autoimmune diseases induced by biological agents. A review of 12,731 cases (BIOGEAS Registry). Expert Opin Drug Saf 2017; 16:1255-1271. [DOI: 10.1080/14740338.2017.1372421] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Marta Pérez-De-Lis
- Servicio de Anestesiologia y Reanimación, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Soledad Retamozo
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Hospital Privado Universitario de Córdoba, Córdoba, Argentina
- Instituto De Investigaciones En Ciencias De La Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Alejandra Flores-Chávez
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Unidad de Investigación Biomédica 02, Unidad de Investigación en Epidemiología Clínica, Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Hospital de Especialidades, Guadalajara, Mexico
- Programa de Doctorado en Ciencias Médicas, Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, Mexico
| | | | | | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA- Sanitas, Barcelona, Spain
| | - Manuel Ramos-Casals
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
68
|
Hegde UP, Mukherji B. Current status of chimeric antigen receptor engineered T cell-based and immune checkpoint blockade-based cancer immunotherapies. Cancer Immunol Immunother 2017; 66:1113-1121. [PMID: 28497159 PMCID: PMC5579167 DOI: 10.1007/s00262-017-2007-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 04/22/2017] [Indexed: 12/27/2022]
Abstract
Adoptive cell therapies with chimeric antigen receptor (CAR) engineered T cells (CAR-T) and immune checkpoint inhibition (ICI)-based cancer immunotherapies have lately shown remarkable success in certain tumor types. CAR-T cell-based therapies targeting CD19 can now induce durable remissions as well as prolong disease-free survival of patients with CD19 positive treatment refractory B cell malignancies and ICI-based therapies with humanized monoclonal antibodies against the T cell inhibitory receptors CTLA-4 and PD-1 as well as against the PD-1 ligand, PD-L1, can now achieve durable remissions as well as prolongation of life of a sizeable fraction of patients with melanoma and Hodgkin's lymphoma and non-small cell cancers. Most importantly, these immuno-therapeutic treatment modalities have raised the possibility of achieving long-term "containment" as well as "cures" for certain types of cancer. While this represents major advances in cancer immunotherapy, both modalities come with considerable toxicities, including fatalities. Although more work will be needed to bring CAR-T cell-based therapies to the bedside for most major cancers and a good deal more will be needed to make ICI-alone or in combination with other treatment modalities-work more consistently and across most major cancers, these two treatment modalities stand out as superb examples of successful translation of bench research to the bedside as well as represent real progress in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Upendra P Hegde
- University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA
| | - Bijay Mukherji
- University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06030, USA.
| |
Collapse
|
69
|
Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immune checkpoint blockers for cancer therapy. Oncoimmunology 2017; 6:e1373237. [PMID: 29147629 DOI: 10.1080/2162402x.2017.1373237] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.
Collapse
Affiliation(s)
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lucillia Bezu
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Kepp
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
70
|
Yin J, Paoletti X, Sargent DJ, Mandrekar SJ. Repeated measures dose-finding design with time-trend detection in the presence of correlated toxicity data. Clin Trials 2017; 14:611-620. [PMID: 28764555 DOI: 10.1177/1740774517723829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Phase I trials are designed to determine the safety, tolerability, and recommended phase 2 dose of therapeutic agents for subsequent testing. The dose-finding paradigm has thus traditionally focused on identifying the maximum tolerable dose of an agent or combination therapy under the assumption that there is a non-decreasing relationship between dose-toxicity and dose-efficacy. The dose is typically determined based on the probability of severe toxicity observed during the first treatment cycle. A novel endpoint, the total toxicity profile, was previously developed to account for the multiple toxicity types and grades experienced in the first cycle. More recently, this was extended to a repeated measures design based on the total toxicity profile to account for longitudinal toxicities over multiple treatment cycles in the absence of within-patient correlation. METHODS In this work, we propose to extend the design in the presence of within-patient correlation. Furthermore, we provide a framework to detect a toxicity time trend (toxicity increasing, decreasing, or stable) over multiple treatment cycles. We utilize a linear mixed model in the Bayesian framework, with the addition of Bayesian risk functions for decision-making in dose assignment. RESULTS The performance of this design was evaluated using simulation studies and real data from a phase I trial. We demonstrated that using available toxicity data from all cycles of treatment improves the accuracy of maximum tolerated dose identification and allows for the detection of a time trend. The performance is consistent regardless of the strength of the within-patient correlation. In addition, the use of a quasi-continuous total toxicity profile score significantly increased the power to detect time trends compared to when binary data only were used. CONCLUSION The increased interest in molecularly targeted agents and immunotherapies in oncology necessitates innovative phase I study designs. Our proposed framework provides a tool to tackle some of the challenges presented by these novel agents, specifically through the ability to understand patterns of toxicity over time, which is important in the cases of cumulative or late toxicities.
Collapse
Affiliation(s)
- Jun Yin
- 1 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Xavier Paoletti
- 2 Biostatistics and Epidemiology Department, INSERM CESP, OncoStat, Institut Gustave Roussy, Villejuif, France
| | - Daniel J Sargent
- 1 Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
71
|
Kyi C, Postow MA. Immune checkpoint inhibitor combinations in solid tumors: opportunities and challenges. Immunotherapy 2017; 8:821-37. [PMID: 27349981 DOI: 10.2217/imt-2016-0002] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The emergence of immune 'checkpoint inhibitors' such as cytotoxic T-lymphocyte antigen 4 (CTLA-4) and programmed death receptor 1 (PD-1) has revolutionized treatment of solid tumors including melanoma, lung cancer, among many others. The goal of checkpoint inhibitor combination therapy is to improve clinical response and minimize toxicities. Rational design of checkpoint combinations considers immune-mediated mechanisms of antitumor activity: immunogenic cell death, antigen release and presentation, activation of T-cell responses, lymphocytic infiltration into tumors and depletion of immunosuppression. Potential synergistic combinations include checkpoint blockade with conventional (radiation, chemotherapy and targeted therapies) and newer immunotherapies (cancer vaccines, oncolytic viruses, among others). Reliable biomarkers are necessary to define patients who will achieve best clinical benefit with minimal toxicity in combination therapy.
Collapse
Affiliation(s)
- Chrisann Kyi
- Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1079, NY 10029, USA
| | - Michael A Postow
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, NY 10065, USA.,Weill Cornell Medical College, 525 E 68th Street, NY 10065, USA
| |
Collapse
|
72
|
Byun DJ, Wolchok JD, Rosenberg LM, Girotra M. Cancer immunotherapy - immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol 2017; 13:195-207. [PMID: 28106152 PMCID: PMC5629093 DOI: 10.1038/nrendo.2016.205] [Citation(s) in RCA: 454] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Advances in cancer therapy in the past few years include the development of medications that modulate immune checkpoint proteins. Cytotoxic T-lymphocyte antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) are two co-inhibitory receptors that are expressed on activated T cells against which therapeutic blocking antibodies have reached routine clinical use. Immune checkpoint blockade can induce inflammatory adverse effects, termed immune-related adverse events (IRAEs), which resemble autoimmune disease. In this Review, we describe the current data regarding immune-related endocrinopathies, including hypophysitis, thyroid dysfunction and diabetes mellitus. We discuss the clinical management of these endocrinopathies within the context of our current understanding of the mechanisms of IRAEs.
Collapse
Affiliation(s)
- David J Byun
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, 10065 New York, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, 10065 New York, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| | - Lynne M Rosenberg
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| | - Monica Girotra
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, 10065 New York, USA
- Weill Cornell Medical College, 1300 York Avenue, New York, 10065 New York, USA
| |
Collapse
|
73
|
Hung AL, Garzon-Muvdi T, Lim M. Biomarkers and Immunotherapeutic Targets in Glioblastoma. World Neurosurg 2017; 102:494-506. [PMID: 28300714 DOI: 10.1016/j.wneu.2017.03.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/28/2017] [Accepted: 03/02/2017] [Indexed: 12/14/2022]
Abstract
Glioblastoma (GBM) is an aggressive central nervous system cancer with poor prognosis despite maximal therapy. The recent advent of immunotherapy holds great promise for improving GBM survival and has already made great strides toward changing management strategies. A diverse set of biomarkers have been implicated as immunotherapeutic targets and prognostic indicators in other cancers. Some of the more extensively studied examples include cytokines (IL-4, IL-13, and TGF-β), checkpoint molecules (PD-1, CTLA-4, TIM-3, LAG-3, CD137, GITR, OX40), and growth/angiogenesis proteins (endoglin and EGFR). Emerging theories involving the tumor mutational landscape and microbiome have also been explored in relation to cancer treatment. Although identification of novel biomarkers may improve and help direct treatment of patients with GBM, the next step is to explore the role of biomarkers in precision medicine and selection of specific immunotherapeutic drugs in an individualized manner.
Collapse
Affiliation(s)
- Alice L Hung
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tomas Garzon-Muvdi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
74
|
Arasanz H, Lacalle A, Lecumberri MJ, de Lascoiti ÁF, Blanco-Luquin I, GatoCañas M, Pérez-Ricarte L, Zuazo M, Kochan G, Escors D. Immunotherapy in malignant melanoma: recent approaches and new perspectives. Melanoma Manag 2017; 4:39-48. [PMID: 30190903 PMCID: PMC6094586 DOI: 10.2217/mmt-2016-0019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 09/07/2016] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy has radically transformed the management of metastatic malignant melanoma. Ipilimumab, a CTLA-4-targeted monoclonal antibody, was the first immunotherapeutic drug to reach a survival benefit compared with traditional chemotherapy. PD-1 targeted therapies, pembrolizumab and nivolumab, have demonstrated, in recent clinical trials, to be even more effective and safer. PD-1 and CTLA-4 blockade combination appears to improve the outcomes achieved so far, although increasing toxicity. However, many questions concerning the optimal timing of administration or the most adequate sequence of treatment are yet to be answered.
Collapse
Affiliation(s)
- Hugo Arasanz
- Medical Oncology Department, Complejo Hospitalario de Navarra, Irunlarrea 3, 31008, Pamplona, Spain
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdisNA. Irunlarrea 3, 31008, Pamplona, Spain
| | - Alejandra Lacalle
- Medical Oncology Department, Complejo Hospitalario de Navarra, Irunlarrea 3, 31008, Pamplona, Spain
| | - Maria José Lecumberri
- Medical Oncology Department, Complejo Hospitalario de Navarra, Irunlarrea 3, 31008, Pamplona, Spain
| | | | - Idoia Blanco-Luquin
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdisNA. Irunlarrea 3, 31008, Pamplona, Spain
| | - María GatoCañas
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdisNA. Irunlarrea 3, 31008, Pamplona, Spain
| | - Leyre Pérez-Ricarte
- Medical Oncology Department, Complejo Hospitalario de Navarra, Irunlarrea 3, 31008, Pamplona, Spain
| | - Miren Zuazo
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdisNA. Irunlarrea 3, 31008, Pamplona, Spain
| | - Grazyna Kochan
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdisNA. Irunlarrea 3, 31008, Pamplona, Spain
| | - David Escors
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdisNA. Irunlarrea 3, 31008, Pamplona, Spain
| |
Collapse
|
75
|
Tanvetyanon T, Gray JE, Antonia SJ. PD-1 checkpoint blockade alone or combined PD-1 and CTLA-4 blockade as immunotherapy for lung cancer? Expert Opin Biol Ther 2017; 17:305-312. [PMID: 28064556 DOI: 10.1080/14712598.2017.1280454] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Signaling through T-cell surface, an immune checkpoint protein such as PD-1 or CTLA-4 helps dampen or terminate unwanted immune responses. Blocking a single immune checkpoint or multiple checkpoints simultaneously can generate anti-tumor activity against a variety of cancers including lung cancer. Area covered: This review highlights the results of recent clinical studies of single or combination checkpoint inhibitor immunotherapy in non-small cell lung cancer (NSCLC) or small cell lung cancer (SCLC). The authors discuss pembrolizumab and pembrolizumab plus ipilimumab, durvalumab and durvalumab plus tremelimumab, nivolumab and nivolumab plus ipilimumab for NSCLC as well as nivolumab and nivolumab plus ipilimumab for SCLC. Expert opinion: Available data suggest that, in both metastatic NSCLC and SCLC, combined PD-1 and CTLA-4 blockade may produce a higher tumor response rate than PD-1 blockade alone. Nevertheless, combination therapy is associated with an increased toxicity. Several larger-scale studies are currently ongoing. For checkpoint inhibitor immunotherapy in SCLC and NSCLC, combination therapy is associated with a higher incidence of toxicities than single therapy; however, it appears to help increase tumor response rate. The increased response rate, if confirmed in larger scale studies, will likely make combination therapy another useful therapeutic approach for lung cancer.
Collapse
Affiliation(s)
- Tawee Tanvetyanon
- a Thoracic Oncology Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Jhanelle E Gray
- a Thoracic Oncology Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| | - Scott J Antonia
- a Thoracic Oncology Department , H. Lee Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| |
Collapse
|
76
|
Cuzzubbo S, Javeri F, Tissier M, Roumi A, Barlog C, Doridam J, Lebbe C, Belin C, Ursu R, Carpentier AF. Neurological adverse events associated with immune checkpoint inhibitors: Review of the literature. Eur J Cancer 2017; 73:1-8. [PMID: 28064139 DOI: 10.1016/j.ejca.2016.12.001] [Citation(s) in RCA: 347] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 12/06/2016] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors (ICIs) targeting CTLA4 and PD1 constitute a promising class of cancer treatment but are associated with several immune-related disorders. We here review the literature reporting neurological adverse events (nAEs) associated with ICIs. A systematic search of literature, up to February 2016, mentioning nAEs in patients treated with ICIs was conducted. Eligible studies included case reports and prospective trials. One case seen in our ward was also added. Within the 59 clinical trials (totalling 9208 patients) analysed, the overall incidence of nAEs was 3.8% with anti-CTLA4 antibodies, 6.1% with anti-PD1 antibodies, and 12.0% with the combination of both. The clinical spectrum of neurological disorders was highly heterogeneous. Most of these nAEs were grade 1-2 and consisted of non-specific symptoms such as headache (55%). The incidence of high grade nAEs was below 1% for all types of treatment. Headaches, encephalopathies and meningitis were the most commonly reported (21%, 19% and 15%, respectively). Among the 27 case reports, the most common nAEs were encephalopathies, meningoradiculoneuritis, Guillain-Barré like syndromes and myasthenic syndromes. The median time of nAEs onset was 6 weeks. In most cases, drug interruption and steroids led to neurological recovery, even in conditions where steroids are not usually recommended such as Guillain-Barré syndrome.
Collapse
Affiliation(s)
- S Cuzzubbo
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France; Université Paris 13, UFR de Santé, Médecine et Biologie Humaine, Bobigny, France.
| | - F Javeri
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France
| | - M Tissier
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France
| | - A Roumi
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France
| | - C Barlog
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France; Université Paris 13, UFR de Santé, Médecine et Biologie Humaine, Bobigny, France
| | - J Doridam
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France; Université Paris 13, UFR de Santé, Médecine et Biologie Humaine, Bobigny, France
| | - C Lebbe
- APHP Dermatology and CIC Departments, INSERM U976, Université Paris Diderot, Sorbonne Paris Cité, Hôpital Saint Louis, Paris, France
| | - C Belin
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France; Université Paris 13, UFR de Santé, Médecine et Biologie Humaine, Bobigny, France
| | - R Ursu
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France; Université Paris 13, UFR de Santé, Médecine et Biologie Humaine, Bobigny, France
| | - A F Carpentier
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Avicenne, Service de Neurologie, Bobigny, France; Université Paris 13, UFR de Santé, Médecine et Biologie Humaine, Bobigny, France
| |
Collapse
|
77
|
CiRen B, Wang X, Long Z. The evaluation of immunotherapy and chemotherapy treatment on melanoma: a network meta-analysis. Oncotarget 2016; 7:81493-81511. [PMID: 27845904 PMCID: PMC5348408 DOI: 10.18632/oncotarget.13277] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/17/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Melanoma is a highly malignant tumor that develops from a neural crest derivative called melanocytes. Chemotherapy is recommended for patients with stage III/IV melanoma. Immunomodulation has also been shown to effectively improve the survival rate of such patients. In the current study, we aimed to perform a network meta-analysis on the therapeutic value of chemotherapy and immunotherapy on melanoma. RESULTS Twenty randomized controlled trials (RCTs) were enrolled in the study. Our Results indicated that ipilimumab + nivolumab had the highest response rate among all therapies, pembrolizumab also had a good efficacy with an excellent tolerance. Chemotherapy had a low response rate, high adverse effects and progressive diseases qualities, therefore it is not recommended as a preferred treatment for patients with advanced melanoma. METHODS The Cochrane library, PubMed and Embase databases were searched for relevant articles. Results of the pair-wise meta-analysis were illustrated by odd ratios (ORs) and corresponding 95% confidence intervals (CIs). Network meta-analysis was performed using a random-effects model under Bayesian framework. Results were illustrated by cumulative ORs and corresponding 95% credible interval (CrIs). The probabilities and outcomes of each treatment were ranked and summarized using the surface under the cumulative ranking curve (SUCRA). CONCLUSIONS We recommend pembrolizumab as the preferred treatment due to its high efficacy and low adverse effects, combination of ipilimumab and nivolumab could be used in severe symptoms.
Collapse
Affiliation(s)
- BaSang CiRen
- Department of Medicine, Shigatse People's Hospital, Shigatse, Tibet, 85700, China
| | - Xinhua Wang
- Department of Dermatology, Shigatse People's Hospital, Shigatse, Tibet, 85700, China
| | - Ziwen Long
- Department of Gastric Cancer and Soft-Tissue Sarcoma Sugery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
78
|
Yang Y, Pei J, Gao G, Yang Z, Guo S, Yue B, Qiu J. Pharmacological interventions for melanoma: Comparative analysis using bayesian meta-analysis. Oncotarget 2016; 7:80855-80871. [PMID: 27764796 PMCID: PMC5348360 DOI: 10.18632/oncotarget.12644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/21/2016] [Indexed: 11/25/2022] Open
Abstract
We conducted a network meta-analysis in order to compare different strategies for managing melanoma patients. Electronic databases were searched for eligible randomized trials that compared different strategies in efficacy and tolerability. Five interventions were associated with a significant improvement in PFS over chemotherapy (all HR < 1): Ipilimumab, Tremelimumab, Nivolumab, Pembrolizumab 2 mg/kg and Ipilimumab + Nivolumab. Three interventions exhibited significantly improved OS results over chemotherapy (all HR < 1): Ipilimumab, Nivolumab and Ipilimumab + Chemotherapy. Four interventions were superior to chemotherapy in CR and PR (all OR > 1): Nivolumab, Pembrolizumab 10 mg/kg, Pembrolizumab 2 mg/kg and Ipilimumab + Nivolumab. However, the other seven interventions were associated with an increased risk of pruritus compared to chemotherapy (all OR > 1). Ipilimumab, Tremelimumab, Ipilimumab + Nivolumab and Ipilimumab + Chemotherapy might result in a higher risk of diarrhea compared to chemotherapy (all OR > 1). Immune checkpoint therapy or combined interventions might be more effective than chemotherapy for managing melanoma patients. However, chemotherapy appears to be more tolerable than these combined strategies with respect to adverse events.
Collapse
Affiliation(s)
- Yang Yang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Medical Insurance and New Rural Cooperative Medical Insurance Administration Center, The 252nd Hospital of PLA, Baoding, 071000, China
| | - Jiaomiao Pei
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Guozhen Gao
- Department of Burn and Plastic Surgery, The 253rd Hospital of PLA, Hohhot, 010051, China
| | - Zheng Yang
- The First Brigade of Fourth Military Medical University, Xi'an, 710032, China
| | - Shuzhong Guo
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Yue
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jianhua Qiu
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
79
|
Ray A, Williams MA, Meek SM, Bowen RC, Grossmann KF, Andtbacka RH, Bowles TL, Hyngstrom JR, Leachman SA, Grossman D, Bowen GM, Holmen SL, VanBrocklin MW, Suneja G, Khong HT. A phase I study of intratumoral ipilimumab and interleukin-2 in patients with advanced melanoma. Oncotarget 2016; 7:64390-64399. [PMID: 27391442 PMCID: PMC5325451 DOI: 10.18632/oncotarget.10453] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 06/25/2016] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Intratumoral interleukin-2 (IL-2) is effective but does not generate systemic immunity. Intravenous ipilimumab produces durable clinical response in a minority of patients, with potentially severe toxicities. Circulating anti-tumor T cells activated by ipilimumab may differ greatly from tumor-infiltrating lymphocytes activated by intratumoral ipilimumab in phenotypes and functionality. The objective of this study was to primarily assess the safety of intratumoral ipilimumab/IL-2 combination and to obtain data on clinical efficacy. RESULTS There was no dose limiting toxicity. While local response of injected lesions was observed in 67% patients (95% CI, 40%-93%), an abscopal response was seen in 89% (95% CI, 68%-100%). The overall response rate and clinical benefit rate by immune-related response criteria (irRC) was 40% (95% CI, 10%-70%) and 50% (95% CI, 19%-81%), respectively. Enhanced systemic immune response was observed in most patients and correlated with clinical responses. EXPERIMENTAL DESIGN Twelve patients with unresectable stages III/IV melanoma were enrolled. A standard 3+3 design was employed to assess highest tolerable intratumoral dose of ipilimumab and IL-2 based on toxicity during the first three weeks. Escalated doses of ipilimumab was injected into only one lesion weekly for eight weeks in cohorts of three patients. A fixed dose of IL-2 was injected three times a week into the same lesion for two weeks, followed by two times a week for six weeks. CONCLUSIONS Intratumoral injection with the combination of ipilimumab/IL-2 is well tolerated and generates responses in both injected and non-injected lesions in the majority of patients.
Collapse
Affiliation(s)
- Abhijit Ray
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | | | - Stephanie M. Meek
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Randy C. Bowen
- School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Kenneth F. Grossmann
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | - Robert H.I. Andtbacka
- Section of Surgical Oncology, Division of General Surgery Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | - Tawnya L. Bowles
- Department of General Surgery, Intermountain Medical Center, Murray, UT, USA
| | - John R. Hyngstrom
- Section of Surgical Oncology, Division of General Surgery Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
- Department of General Surgery, Intermountain Medical Center, Murray, UT, USA
| | - Sancy A. Leachman
- Department of Dermatology, Oregon Health & Science University-Knight Cancer Institute, Portland, OR, USA
| | - Douglas Grossman
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | - Glen M. Bowen
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | - Sheri L. Holmen
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | - Matthew W. VanBrocklin
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| | - Gita Suneja
- Department of Radiation Oncology, University of Utah, Salt Lake City, UT, USA
| | - Hung T. Khong
- Division of Oncology, Huntsman Cancer Institute-University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
80
|
González-Rodríguez E, Rodríguez-Abreu D. Immune Checkpoint Inhibitors: Review and Management of Endocrine Adverse Events. Oncologist 2016; 21:804-16. [PMID: 27306911 PMCID: PMC4943391 DOI: 10.1634/theoncologist.2015-0509] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 02/19/2016] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED : In recent years, immune checkpoint inhibitors have emerged as effective therapies for advanced neoplasias. As new checkpoint target blockers become available and additional tumor locations tested, their use is expected to increase within a short time. Immune-related adverse events (irAEs) affecting the endocrine system are among the most frequent and complex toxicities. Some may be life-threatening if not recognized; hence, appropriate guidance for oncologists is needed. Despite their high incidence, endocrine irAEs have not been fully described for all immunotherapy agents available. This article is a narrative review of endocrinopathies associated with cytotoxic T lymphocyte-associated antigen-4, blockade of programmed death receptor 1 and its ligand inhibitors, and their combination. Thyroid dysfunction is the most frequent irAE reported, and hypophysitis is characteristic of ipilimumab. Incidence, timing patterns, and clinical presentation are discussed, and practical recommendations for clinical management are suggested. Heterogeneous terminology and lack of appropriate resolution criteria in clinical trials make adequate evaluation of endocrine AEs difficult. It is necessary to standardize definitions to contrast incidences and characterize toxicity patterns. To provide optimal care, a multidisciplinary team that includes endocrinology specialists is recommended. IMPLICATIONS FOR PRACTICE Immune checkpoint inhibitors are already part of oncologists' therapeutic arsenal as effective therapies for otherwise untreatable neoplasias, such as metastatic melanoma or lung cancer. Their use is expected to increase exponentially in the near future as additional agents become available and their approval is extended to different tumor types. Adverse events affecting the endocrine system are among the most frequent and complex toxicities oncologists may face, and some may be life-threatening if not recognized. This study reviews endocrinopathies associated to immune checkpoint inhibitors available to date. Incidence, timing patterns, and clinical presentation are discussed, and practical recommendations for management are proposed.
Collapse
Affiliation(s)
- Elisa González-Rodríguez
- Section of Endocrinology and Nutrition, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canaria, Spain
| | - Delvys Rodríguez-Abreu
- Section of Medical Oncology, Hospital Universitario Insular de Gran Canaria, Las Palmas de Gran Canaria, Spain
| |
Collapse
|
81
|
Petrelli F, Coinu A, Cabiddu M, Borgonovo K, Ghilardi M, Lonati V, Barni S. Early analysis of surrogate endpoints for metastatic melanoma in immune checkpoint inhibitor trials. Medicine (Baltimore) 2016; 95:e3997. [PMID: 27368007 PMCID: PMC4937921 DOI: 10.1097/md.0000000000003997] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Recent major phase III trials led to the approval of immune checkpoint inhibitors (ipilimumab, pembrolizumab, and nivolumab) in metastatic malignant melanoma (MM). We aim to assess whether median progression-free survival, and 1 and 2-year overall survival (OS) rates are reliable surrogate endpoints for median OS through a meta-analysis of published trials involving immunotherapy. A systematic literature search in PubMed, EMBASE, Web of Science, and SCOPUS of published phase II to III trials with immunotherapy as the treatment for MM was conducted. Adjusted weighted linear regression was used to calculate Pearson correlations (R) between surrogates and median OS, and between treatment effects on surrogates and median OS. A total of 13 studies involving 3373 patients with MM were identified. The correlation of progression-free survival with OS was not significant (R = 0.45, P = .11). Conversely, the correlation between 1-year OS and median OS was very strong (R = 0.93, 95% confidence interval [CI] 0.84-0.96, P < .00001), as was the correlation between 2-year OS and OS (R = 0.79, 95% CI 0.51-0.91, P = .0001). The correlation between the treatment effects on 1-year OS and OS was also significant (R = -0.86, 95% CI -0.3 to 0.97, P = .01). Similar results were obtained for 2-year OS. According to the available study data, 1-year OS rate could be regarded as a potential surrogate for median OS in novel immunotherapy trials of metastatic MM. Waiting for ongoing studies (e.g., pembrolizumab), we suggest that this intermediate endpoint could be considered as a potential primary endpoint in future clinical trials.
Collapse
Affiliation(s)
- Fausto Petrelli
- Oncology Department, Oncology Division, ASST Bergamo Ovest, Treviglio (BG), Italy
- Correspondence: Fausto Petrelli, Oncology Department, Oncology Division, ASST Bergamo Ovest, Piazzale Ospedale 1, 24047 Treviglio (BG), Italy (e-mail: )
| | | | | | | | | | | | | |
Collapse
|
82
|
Safety profiles of anti-CTLA-4 and anti-PD-1 antibodies alone and in combination. Nat Rev Clin Oncol 2016; 13:473-86. [DOI: 10.1038/nrclinonc.2016.58] [Citation(s) in RCA: 660] [Impact Index Per Article: 82.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
83
|
Abstract
Similar to cutaneous melanoma, several strategies of immune escape have been documented in uveal melanomas (UMs). We hypothesized that these cancers could respond to cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) inhibition with tremelimumab by potentiating T-cell activation. This was an open-label, multicentre phase 2 study in patients with advanced UM who had not received prior immunotherapy. Patient received tremelimumab at 15 mg/kg administered every 90 days for up to four cycles. The primary endpoint was 6-month progression-free survival (PFS). Secondary endpoints were safety, durable response rate, objective response rate, duration of objective response, duration of complete response, and median overall survival (OS). Eleven patients, all with M1c disease, were enrolled with no responses observed. The median follow-up was 11 months (range 2-36 months). The median PFS was 2.9 months (95% confidence interval 2.8-3.0) and the 6-month PFS rate was 9.1%. The median OS was 12.8 months (95% confidence interval 3.8-19.7). Toxicities were consistent with CTLA-4 blockade and were manageable. Although the median OS of 12.8 months and the manageable toxicity profile of tremelimumab observed in this study seem promising, the modest 6-month PFS and the lack of responses observed resulted in the study being stopped due to futility at the first interim stage. To date, no systemic treatment has demonstrated a survival benefit in patients with advanced UM. The standard treatment for patients with advanced UM should be a clinical trial.
Collapse
|
84
|
Comin-Anduix B, Escuin-Ordinas H, Ibarrondo FJ. Tremelimumab: research and clinical development. Onco Targets Ther 2016; 9:1767-76. [PMID: 27042127 PMCID: PMC4809326 DOI: 10.2147/ott.s65802] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The immune checkpoint therapy is a relatively recent strategy that aims to tweak the immune system to effectively attack cancer cells. The understanding of the immune responses and their regulation at the intracellular level and the development of fully humanized monoclonal antibodies are the pillars of an approach that could elicit durable clinical responses and even remission in some patients with cancer. Most of the immune checkpoints that regulate the T-cell responses (activation and inhibition) operate through proteins present on the cytoplasmic membrane of the immune cells. Therefore, specific antibodies capable of blocking the inhibitory signals should lead to unrestrained immune responses that supersede the inhibitory mechanisms, which are naturally present in the tumor microenviroment. The best-known and most successful targets for immune checkpoint therapy are the cytotoxic T-lymphocyte antigen-4 and programmed cell death-1 coreceptors. Tremelimumab (CP-675,206) is a fully humanized monoclonal antibody specific for cytotoxic T-lymphocyte antigen-4, which has been successfully used to treat patients with metastatic melanoma and some other cancers. Although still a work in progress, the use of tremelimumab as an immune checkpoint therapeutic agent is a promising approach alone or in combination with other anticancer drugs. Here, we review the use of this antibody in a number of clinical trials against solid tumors.
Collapse
Affiliation(s)
- Begoña Comin-Anduix
- Division of Surgical-Oncology, Department of Surgery, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Helena Escuin-Ordinas
- Division of Hematology-Oncology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Francisco Javier Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
85
|
Abstract
The immune system is capable of recognizing tumors and eliminates many early malignant cells. However, tumors evolve to evade immune attack, and the tumor microenvironment is immunosuppressive. Immune responses are regulated by a number of immunological checkpoints that promote protective immunity and maintain tolerance. T cell coinhibitory pathways restrict the strength and duration of immune responses, thereby limiting immune-mediated tissue damage, controlling resolution of inflammation, and maintaining tolerance to prevent autoimmunity. Tumors exploit these coinhibitory pathways to evade immune eradication. Blockade of the PD-1 and CTLA-4 checkpoints is proving to be an effective and durable cancer immunotherapy in a subset of patients with a variety of tumor types, and additional combinations are further improving response rates. In this review we discuss the immunoregulatory functions of coinhibitory pathways and their translation to effective immunotherapies for cancer.
Collapse
Affiliation(s)
- Susanne H Baumeister
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Division of Pediatric Hematology-Oncology, Boston Children's Hospital, Boston, Massachusetts 02115.,Harvard Medical School, Boston, Massachusetts 02115
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Harvard Medical School, Boston, Massachusetts 02115
| | - Glenn Dranoff
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215.,Novartis Institutes for BioMedical Research, Exploratory Immuno-oncology, Cambridge, Massachusetts 02139
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, and Evergrande Center for Immunologic Diseases, Harvard Medical School, Boston, Massachusetts 02115;
| |
Collapse
|
86
|
Abstract
In recent years, the introduction and Federal Drug Administration approval of immune checkpoint inhibitor antibodies has dramatically improved the clinical outcomes for patients with advanced melanoma. These antagonist monoclonal antibodies are capable of unleashing dormant or exhausted antitumor immunity, which has led to durable complete and partial responses in a large number of patients. Ipilimumab targets the cytotoxic T lymphocyte-associated protein 4 (CTLA-4) receptor. Nivolumab and pembrolizumab target programmed cell death protein 1 (PD-1) receptors and have proven to be superior to ipilimumab alone. The combination of ipilimumab and nivolumab has yielded higher response rates, greater tumor shrinkage, and longer progression-free survival than either monotherapy alone. As other promising immunotherapies for melanoma proceed through clinical trials, future goals include defining the role of immune checkpoint inhibitors as adjuvant therapy, identifying optimal combination strategies, and developing reliable predictive biomarkers to guide treatment selection for individual patients.
Collapse
Affiliation(s)
- Jason M Redman
- Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007, Washington DC, USA. .,Department of Medicine, Georgetown University Medical Center, Washington DC, USA.
| | - Geoffrey T Gibney
- Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007, Washington DC, USA. .,Department of Medicine, Georgetown University Medical Center, Washington DC, USA.
| | - Michael B Atkins
- Georgetown Lombardi Comprehensive Cancer Center 3970 Reservoir Road, NW Research Building, Room E501, 20007, Washington DC, USA. .,Department of Medicine, Georgetown University Medical Center, Washington DC, USA.
| |
Collapse
|
87
|
Guazzelli A, Hussain M, Krstic-Demonacos M, Mutti L. Tremelimumab for the treatment of malignant mesothelioma. Expert Opin Biol Ther 2015; 15:1819-29. [DOI: 10.1517/14712598.2015.1116515] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
88
|
Márquez-Rodas I, Cerezuela P, Soria A, Berrocal A, Riso A, González-Cao M, Martín-Algarra S. Immune checkpoint inhibitors: therapeutic advances in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:267. [PMID: 26605313 DOI: 10.3978/j.issn.2305-5839.2015.10.27] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In recent years, new strategies for treating melanoma have been introduced, improving the outlook for this challenging disease. One of the most important advances has been the development of immunotherapy. The better understanding of the role of the immunological system in tumor control has paved the way for strategies to enhance the immune response against cancer cells. Monoclonal antibodies (mAbs) against the immune checkpoints cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have demonstrated high activity in melanoma and other tumors. Ipilimumab, an anti CTLA-4 antibody, was the first drug of this class that was approved. Although the response rate with ipilimumab is low (less than 20% of patients have objective responses), 20% of patients have long survival, with similar results in the first and second line settings. Nivolumab and pembrolizumab, both anti PD-1 inhibitors, have been approved for the treatment of melanoma, with response rates of 40% and a demonstrated survival advantage in phase III trials. This has marked a new era in the treatment of metastatic melanoma and much research is now ongoing with other drugs targeting checkpoint inhibitors. In addition, the agonist of activating molecules on T cells and their combinations are being investigated. Herein we review the clinical development of checkpoint inhibitors and their approval for treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Ivan Márquez-Rodas
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Pablo Cerezuela
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Ainara Soria
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Alfonso Berrocal
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Aldo Riso
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - María González-Cao
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| | - Salvador Martín-Algarra
- 1 Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañon, Madrid, Spain ; 2 Spanish Melanoma Group (GEM); 3 Servicio Oncología Médica, Hospital General Universitario Santa Lucía, Cartagena, Spain ; 4 Servicio de Oncología Médica, Universitario Ramon y Cajal, Madrid, Spain ; 5 Servicio de Oncología Médica, Hospital General Universitario de Valencia, Valencia, Spain ; 6 Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain ; 7 Servicio de Oncología Médica, Clínica Universitaria de Navarra, Pamplona, Spain
| |
Collapse
|
89
|
Postel-Vinay S, Aspeslagh S, Lanoy E, Robert C, Soria JC, Marabelle A. Challenges of phase 1 clinical trials evaluating immune checkpoint-targeted antibodies. Ann Oncol 2015; 27:214-24. [PMID: 26578728 DOI: 10.1093/annonc/mdv550] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Immunostimulatory monoclonal antibodies (imAbs) targeting immune checkpoint molecules are revolutionizing oncology not only regarding cancer therapeutics and clinical care, but also from a drug development point of view. A handful of first-generation molecules have been approved so far based on their tremendous efficacy, after an expedited development phase that has challenged most paradigms established in the era of conventional cytotoxic therapy and to some extent molecularly targeted agents. A huge wave of second-generation imAbs is just entering into phase 1 trials now, in monotherapy or in combination. In order to maximize their chances of success in early phase trials, and eventually for patients' benefit, their clinical development has to benefit from lessons learnt from previous imAbs phase 1 trials. MATERIALS AND METHODS We reviewed the early clinical development of anti-cytotoxic T-lymphocyte antigen 4 and anti-programmed death-1 receptor/ligand. Particularities of each agent, including safety, dose--toxicity and dose--efficacy relationships, scheduling, pharmacokinetics (PK), pharmacodynamics (PD), trial design, biomarkers, response assessment and overall drug development strategies, are described and challenged. RESULTS As opposed to conventional cytotoxic agents, dose of imAbs is not linearly associated with efficacy and toxicity. Therefore, the definition of a minimal immunologically active dose could be proposed. Traditional patient eligibility criteria might also be revisited as the toxicity profile and mechanism of toxicity--immune-related adverse events--are mostly known and their physiopathology somehow less unexpected than with molecularly targeted small molecules. Most challenging are the comprehensive investigation of complex PK and PD characteristics as well as the definition of patient selection biomarkers. Finally, the early focus on efficacy (and not only dose confirmation) in expansion cohorts challenges the traditional phase 1/2/3 drug development process. CONCLUSION Several drug development paradigms have been challenged by imAbs. Here, we discuss novel approaches for an efficient and successful drug development of these agents.
Collapse
Affiliation(s)
- S Postel-Vinay
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - S Aspeslagh
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif
| | - E Lanoy
- Biostatistics and Epidemiology Unit, Gustave-Roussy, Villejuif Inserm Unit U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif
| | - C Robert
- Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif Department of Medical Oncology, Gustave Roussy, Villejuif
| | - J-C Soria
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Faculty of Medicine, Université Paris Saclay, Université Paris-Sud, Paris Inserm Unit U981, Gustave Roussy, Villejuif
| | - A Marabelle
- DITEP (Département d'Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif Inserm Unit U1015, Gustave Roussy, Villejuif, France
| |
Collapse
|
90
|
Eroglu Z, Kim DW, Wang X, Camacho LH, Chmielowski B, Seja E, Villanueva A, Ruchalski K, Glaspy JA, Kim KB, Hwu WJ, Ribas A. Long term survival with cytotoxic T lymphocyte-associated antigen 4 blockade using tremelimumab. Eur J Cancer 2015; 51:2689-97. [PMID: 26364516 PMCID: PMC4821004 DOI: 10.1016/j.ejca.2015.08.012] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 06/25/2015] [Accepted: 08/14/2015] [Indexed: 11/23/2022]
Abstract
PURPOSE One of the hallmarks of cancer immunotherapy is the long duration of responses, evident with cytokines like interleukin-2 or a variety of cancer vaccines. However, there is limited information available on very long term outcomes of patients treated with anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies. Tremelimumab is an anti-CTLA-4 antibody of immunoglobulin G2 (IgG2) isotype initially tested in patients with advanced melanoma over 12 years ago. METHODS We reviewed the outcomes of patients with advanced melanoma enrolled in four phase 1 and 2 tremelimumab trials at two sites to determine response rates and long-term survival. RESULTS A total of 143 patients were enrolled at two institutions from 2002 to 2008. Tremelimumab administration varied between a single dose of 0.01 mg/kg and 15 mg/kg every 3 months. Median overall survival was 13 months (95% confidence interval (CI), 10-16.6), ranging from less than a month to 12+ years. An objective response rate of 15.6% was observed, with median duration of response of 6.5 years, range of 3-136+ months. The Kaplan-Meier estimated 5 year survival rate was 20% (95% CI, 13-26%), with 10 and 12.5 year survival rates of 16% (95% CI, 9-23%). CONCLUSIONS CTLA-4 blockade with tremelimumab can lead to very long duration of objective anti-tumour responses beyond 12 years.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/immunology
- Antineoplastic Agents/therapeutic use
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Immunotherapy/methods
- Kaplan-Meier Estimate
- Male
- Melanoma/immunology
- Melanoma/mortality
- Melanoma/therapy
- Middle Aged
- Outcome Assessment, Health Care/methods
- Outcome Assessment, Health Care/statistics & numerical data
- Remission Induction
- Retrospective Studies
- Survival Rate
- Survivors/statistics & numerical data
- Time Factors
- Young Adult
Collapse
Affiliation(s)
- Zeynep Eroglu
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Dae Won Kim
- Department of Melanoma Medical Oncology, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoyan Wang
- Department of Medicine Statistics Core, University of California Los Angeles, Los Angeles, CA, USA
| | - Luis H Camacho
- St. Luke's Medical Center Cancer Center, Houston, TX, USA
| | - Bartosz Chmielowski
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Elizabeth Seja
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Arturo Villanueva
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Kathleen Ruchalski
- Department of Radiology, University of California Los Angeles, Los Angeles, CA, USA
| | - John A Glaspy
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA
| | - Kevin B Kim
- Department of Melanoma Medical Oncology, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA; California Pacific Medical Center, San Francisco, CA, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, The University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology/Oncology, Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, CA, USA.
| |
Collapse
|
91
|
Teixidó C, González-Cao M, Karachaliou N, Rosell R. Predictive factors for immunotherapy in melanoma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:208. [PMID: 26488004 DOI: 10.3978/j.issn.2305-5839.2015.05.07] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Immunotherapy has emerged as an exciting strategy for cancer treatment. Therapeutic blockade of immune checkpoint regulators favors the ability of T cell responses to increase anti-tumor immunity. The cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed cell death-1 (PD-1) are two T cell-inhibitory receptors with independent mechanisms of action. Immune checkpoint inhibitors targeting either CTLA-4, PD-1 or its ligand PD-L1 are currently yielding promising results in terms of efficacy in several clinical studies with melanoma patients and are being developed and tested as immunotherapy agents for multiple cancer types. To date, no reliable predictors of activity and efficacy of immunotherapy have yet been identified or validated. Even so, determining which patients derive clinical benefit from immune checkpoint agents remains an important clinical question and efforts to identify predictive markers of response are ongoing. This article reviews the current potential predictive factors for CTLA-4 and PD-1/PD-L1 immune checkpoints inhibitors in melanoma.
Collapse
Affiliation(s)
- Cristina Teixidó
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Maria González-Cao
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Niki Karachaliou
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| | - Rafael Rosell
- 1 Pangaea Biotech, 2 Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, 08028 Barcelona, Spain ; 3 Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, 08916 Badalona, Spain ; 4 Molecular Oncology Research Foundation (MORe), Barcelona, Spain
| |
Collapse
|
92
|
Derer A, Deloch L, Rubner Y, Fietkau R, Frey B, Gaipl US. Radio-Immunotherapy-Induced Immunogenic Cancer Cells as Basis for Induction of Systemic Anti-Tumor Immune Responses - Pre-Clinical Evidence and Ongoing Clinical Applications. Front Immunol 2015; 6:505. [PMID: 26500646 PMCID: PMC4597129 DOI: 10.3389/fimmu.2015.00505] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/16/2015] [Indexed: 01/18/2023] Open
Abstract
Radiotherapy (RT) primarily aims to locally destroy the tumor via the induction of DNA damage in the tumor cells. However, the so-called abscopal, namely systemic and immune–mediated, effects of RT move over more and more in the focus of scientists and clinicians since combinations of local irradiation with immune therapy have been demonstrated to induce anti-tumor immunity. We here summarize changes of the phenotype and microenvironment of tumor cells after exposure to irradiation, chemotherapeutic agents, and immune modulating agents rendering the tumor more immunogenic. The impact of therapy-modified tumor cells and damage-associated molecular patterns on local and systemic control of the primary tumor, recurrent tumors, and metastases will be outlined. Finally, clinical studies affirming the bench-side findings of interactions and synergies of radiation therapy and immunotherapy will be discussed. Focus is set on combination of radio(chemo)therapy (RCT) with immune checkpoint inhibitors, growth factor inhibitors, and chimeric antigen receptor T-cell therapy. Well-deliberated combination of RCT with selected immune therapies and growth factor inhibitors bear the great potential to further improve anti-cancer therapies.
Collapse
Affiliation(s)
- Anja Derer
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Lisa Deloch
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Yvonne Rubner
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen , Germany
| |
Collapse
|
93
|
Shimomura A, Fujiwara Y, Kondo S, Kodaira M, Iwasa S, Kitano S, Tanabe Y, Tamura K, Yamamoto N. Tremelimumab-associated tumor regression following after initial progression: two case reports. Immunotherapy 2015; 8:9-15. [PMID: 26427600 DOI: 10.2217/imt.15.89] [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: 01/22/2023] Open
Abstract
The human IgG2 monoclonal antibody tremelimumab is an immune checkpoint inhibitor that blocks cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). The therapeutic response of anti-CTLA-4 monoclonal antibodies possess unique kinetics, in that antitumor responses are often observed after initial short-term disease progression, in some cases as long as 6-12 months after anti-CTLA-4 treatment initiation. Here, we report two cases: one of bile duct cancer and the other of squamous cell carcinoma of unknown primary, both of which demonstrated initial rapid disease progression followed by dramatic tumor shrinkage after one or two doses of tremelimumab, without any immune-related adverse events. This delayed, yet dramatic antitumor response suggests that tremelimumab may hold promise in the treatment of solid tumors.
Collapse
Affiliation(s)
- Akihiko Shimomura
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Department of Breast & Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yutaka Fujiwara
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Makoto Kodaira
- Department of Breast & Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Satoru Iwasa
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigehisa Kitano
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yuko Tanabe
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kenji Tamura
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.,Department of Breast & Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
94
|
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
|
95
|
Loirat D, Le Tourneau C. Immunothérapie anticancer : les molécules immunomodulatrices en développement clinique. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
96
|
Ward FJ, Dahal LN, Khanolkar RC, Shankar SP, Barker RN. Targeting the alternatively spliced soluble isoform of CTLA-4: prospects for immunotherapy? Immunotherapy 2015; 6:1073-84. [PMID: 25428646 DOI: 10.2217/imt.14.73] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
CTLA-4 is an inhibitory protein that contributes to immune homeostasis and tolerance, a role that has led to its exploitation as a therapeutic in several clinical settings including cancer and autoimmune disease. Development of CTLA-4 therapies focused largely on the full-length receptor isoform but other CTLA-4 isoforms are also expressed, including a secretable form of CTLA-4 (soluble CTLA-4 [sCTLA-4]). The contribution of sCTLA-4 to immune regulation has been less well studied, primarily because it was identified some years after the original description of CTLA-4. Here, we examine how sCTLA-4 might contribute to immune regulation and ask whether it might be a biomarker to inform current CTLA-4 therapies or represent a novel CTLA-4 target for future therapeutics.
Collapse
Affiliation(s)
- Frank J Ward
- Section of Immunology & Infection, Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, UK
| | | | | | | | | |
Collapse
|
97
|
Abstract
The potential to harness the power of the immune system and effectively treat patients with metastatic melanoma is finally being realized with the advent of immune checkpoint inhibitors. These new therapies herald a new era in the treatment of melanoma with the potential to produce very durable responses and possible cure for a subset of patients, though bring with them challenges including novel toxicities and nonconventional response patterns. This article reviews the currently available immune checkpoint inhibitors, potential biomarkers to predict response and promising investigational approaches including combination therapies.
Collapse
Affiliation(s)
- Adam J Cooper
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
- School of Medicine, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
- School of Medicine, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
- Westmead Institute for Cancer Research, University of Sydney at the Westmead Millennium Institute, Westmead, NSW, Australia
| | - Richard F Kefford
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
- School of Medicine, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
- Westmead Institute for Cancer Research, University of Sydney at the Westmead Millennium Institute, Westmead, NSW, Australia
- Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
98
|
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]
|
99
|
Marzuka A, Huang L, Theodosakis N, Bosenberg M. Melanoma Treatments: Advances and Mechanisms. J Cell Physiol 2015; 230:2626-33. [DOI: 10.1002/jcp.25019] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Alexander Marzuka
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Internal Medicine; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Laura Huang
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Internal Medicine; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Nicholas Theodosakis
- Department of Pathology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Marcus Bosenberg
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Pathology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| |
Collapse
|
100
|
Abstract
PURPOSE OF REVIEW Over the last 18 months, substantial progress has been made in demonstrating the clinical efficacy of harnessing the immune system to treat a variety of both solid and hematologic malignancies. This review summarizes and evaluates these seminal studies. RECENT FINDINGS The two treatment modalities most responsible for the success of immune based therapies in cancer are adoptive T-cell therapy and immunoregulatory antibodies. Specifically, immunotherapy is generating responses in malignancies that would otherwise have no traditional curative options such as CD19-targeted chimeric antigen receptors to treat relapsed/refractory acute lymphocytic leukemia and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and PD-1 blockade alone or in combination to treat metastatic melanoma and other solid tumors. SUMMARY We are at a turning point for the field of cancer immunotherapy. The scientific community is now, after decades of research, proving that these treatments have great promise for patients. Ongoing preclinical research and clinical trials over the next few years will determine the extent of impact cancer immunotherapy will have on the treatment of the general population.
Collapse
|