301
|
Hao MZ, Zhou WY, Du XL, Chen KX, Wang GW, Yang Y, Yang JL. Novel anti-melanoma treatment: focus on immunotherapy. CHINESE JOURNAL OF CANCER 2015; 33:458-65. [PMID: 25189718 PMCID: PMC4190435 DOI: 10.5732/cjc.014.10118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Melanoma is an intractable cancer that is aggressive, lethal, and metastatic. The prognosis of advanced melanoma is very poor because it is insensitive to chemotherapy and radiotherapy. The incidence of melanoma has been ascending stably for years worldwide, accompanied by increasing mortality. New approaches to managing this deadly disease are much anticipated to enhance the cure rate and to extend clinical benefits to patients with metastatic melanoma. Due to its high degree of immunogenicity, melanoma could be a good target for immunotherapy, which has been developed for decades and has achieved certain progress. This article provides an overview of immunotherapy for melanoma.
Collapse
Affiliation(s)
- Meng-Ze Hao
- Department of Bone and Soft Tissue Tumor, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Hospital & Institute, Tianjin 30060, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
302
|
Yousaf N, Davidson M, Goode E, Thomas C, Hung R, Gore M, Larkin J. The cost of ipilimumab toxicity: a single-centre analysis. Melanoma Res 2015; 25:259-64. [PMID: 25860328 DOI: 10.1097/cmr.0000000000000158] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
There are no published data on the costs associated with investigating and managing toxicity from ipilimumab treatment in patients with metastatic melanoma. Patients treated with ipilimumab at The Royal Marsden Hospital between 1 September 2010 and 1 April 2013 were identified. Data on demographics, investigations and survival outcomes were collected. Patients with grade 3 or higher immune-related adverse events were identified, and costs of investigating and managing toxicities in them were calculated on the basis of standard National Health Service tariffs. Out of the 110 patients, 29 experienced grade 3/4 immune-related adverse events. The total cost of investigating and managing these patients was £140,680, or a median cost of £2860 per patient. Patients experiencing grade 3/4 toxicities had 1-, 2- and 3-year survival rates of 79, 59 and 46%, compared with 24, 17 and 15% in the group that did not experience significant toxicity (P<0.0005). The most common treatment-related toxicity identified was colitis. Two patients died from complications associated with ipilimumab colitis. The cost of ipilimumab toxicity is marginal in comparison with the total treatment cost. Patients treated with ipilimumab who develop significant toxicity have a higher than expected 1-, 2- and 3-year survival.
Collapse
Affiliation(s)
- Nadia Yousaf
- Department of Medicine, Royal Marsden Hospital, London, UK
| | | | | | | | | | | | | |
Collapse
|
303
|
Mozzillo N, Simeone E, Benedetto L, Curvietto M, Giannarelli D, Gentilcore G, Camerlingo R, Capone M, Madonna G, Festino L, Caracò C, Di Monta G, Marone U, Di Marzo M, Grimaldi AM, Mori S, Ciliberto G, Ascierto PA. Assessing a novel immuno-oncology-based combination therapy: Ipilimumab plus electrochemotherapy. Oncoimmunology 2015; 4:e1008842. [PMID: 26155423 PMCID: PMC4485758 DOI: 10.1080/2162402x.2015.1008842] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/12/2015] [Accepted: 01/14/2015] [Indexed: 01/05/2023] Open
Abstract
Melanoma is responsible for most skin cancer-related deaths and is one of the most common cancers diagnosed in young adults. In melanoma, tumors can become established by activation of the negative regulator of cytotoxic T lymphocytes (CTLs), CTL antigen-4 (CTLA-4). Ipilimumab blocks the interaction of CTLA-4 with CD80/CD86 and augments T-cell activation and proliferation. In electrochemotherapy (ECT), local application of short high-voltage pulses renders cell membranes transiently permeable to chemotherapeutic drugs. The combination of ipilimumab and ECT may be beneficial for the treatment of metastatic melanoma; however, no prospective data are available to date. Here, we report the retrospective analysis of patients treated with ipilimumab in an expanded access program (EAP) who also received ECT. Fifteen patients with previously treated metastatic melanoma who received ipilimumab 3 mg/kg every three weeks for four cycles and underwent ECT for local disease control and/or palliation of cutaneous lesions with bleomycin 15 mg/m2 after the first ipilimumab infusion were included in the analysis. Over the study period, a local objective response was observed in 67% of patients (27% complete response [CR] and 40% partial response [PR]). According to immune-related response criteria, a systemic response was observed in nine patients (five PR and four stable disease [SD]), resulting in a disease control rate of 60%. Evaluation of circulating T-regulatory (T-reg) cells demonstrated significant differences between responders and non-responders. Overall, treatment was well-tolerated and without notable toxicity. In conclusion, the combination of ipilimumab and ECT appears to be beneficial to patients with advanced melanoma, warranting further investigation in prospective trials.
Collapse
Affiliation(s)
- Nicola Mozzillo
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Ester Simeone
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Lucia Benedetto
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Marcello Curvietto
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Diana Giannarelli
- Statistical Unit; Regina Elena National Cancer Institute; Rome, Italy
| | - Giusy Gentilcore
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Rosa Camerlingo
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Lucia Festino
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Corrado Caracò
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Gianluca Di Monta
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Ugo Marone
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Massimiliano Di Marzo
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Antonio M Grimaldi
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Stefano Mori
- Melanoma and Sarcoma Surgery Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Gennaro Ciliberto
- Scientific Direction; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy, and Innovative Therapy Unit; Istituto Nazionale Tumori Fondazione “G. Pascale”; Naples, Italy
| |
Collapse
|
304
|
Barbee MS, Ogunniyi A, Horvat TZ, Dang TO. Current status and future directions of the immune checkpoint inhibitors ipilimumab, pembrolizumab, and nivolumab in oncology. Ann Pharmacother 2015; 49:907-37. [PMID: 25991832 DOI: 10.1177/1060028015586218] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE To provide the clinician with an update and the current status and future direction of approved immune checkpoint inhibitors (ICIs) in oncology. DATA SOURCES A PubMed search from January 1, 1966 to March 13, 2015 was performed using the key terms ipilimumab, pembrolizumab, lambrolizumab, nivolumab, immune checkpoint inhibitor, MDX-010, MDX-101, BMS-734016, MK-3475, SCH 900475, MDX-1106, BMS-936558, ONO-4538, CTLA-4, PD-1, or PD-L1 and cancer, oncology, or neoplasm. Additional references were identified from the investigators(') personal files, recent oncology meetings, review articles, clinical guidelines, and package inserts. STUDY SELECTION AND DATA EXTRACTION All English-language clinical trials assessing the safety and efficacy of ipilimumab, nivolumab, and pembrolizumab in cancer were considered. The PubMed search resulted in 215 trials; 33 met inclusion criteria. A further 28 trials were identified from the above sources; 61 trials from 2005 to 2015 were included. We consolidated and clarified treatment recommendations for the management of immune-related adverse events (irAEs), assessed response criteria, and calculated the clinical utility of leading tumor profiling options. DATA SYNTHESIS Ipilimumab and nivolumab, but not pembrolizumab, have an overall survival (OS) advantage over chemotherapy first line in unresectable/metastatic melanoma. Nivolumab has an OS advantage versus chemotherapy in second-line squamous non-small-cell lung cancer. Data in other settings are promising. Nivolumab and pembrolizumab are better tolerated than ipilimumab. Further validation of response criteria is needed. Tumor profiling to predict clinical benefit is premature but promising. CONCLUSIONS The treatment landscape in oncology is quickly evolving with the advent of ICIs.
Collapse
Affiliation(s)
| | | | - Troy Z Horvat
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thu-Oanh Dang
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
305
|
Cheng R, Cooper A, Kench J, Watson G, Bye W, McNeil C, Shackel N. Ipilimumab-induced toxicities and the gastroenterologist. J Gastroenterol Hepatol 2015; 30:657-66. [PMID: 25641691 DOI: 10.1111/jgh.12888] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/29/2014] [Indexed: 12/13/2022]
Abstract
Ipilimumab has been shown to improve overall survival in patients with advanced melanoma. Ipilimumab acts through immune-modulation, and is recognized to cause potentially severe immune-related adverse events (irAEs) including dermatitis, colitis, thyroiditis, hypophysitis, and hepatitis. The acceptance of ipilimumab as a treatment for metastatic melanoma means patients will continue to be treated with this agent and gastroenterologists will be increasingly called upon to assist in managing severe autoimmune-related hepatitis and colitis. To date, the recommendations for managing irAEs secondary to ipilimumab have been steroids at a moderate dose of prednisolone (1 mg/kg) as well as immunosuppressive agents such as mycophenolate mofetil (MMF) for steroid-refractory hepatitis and infliximab in the management of corticosteroid-refractory colitis. However, the dosing and the duration of immunosuppressive therapy have not been systematically studied in the setting of treating ipilimumab-induced irAEs. Therefore, additional immune-modifying agents and/or a change in dosing may be required to manage severe irAEs unresponsive to existing treatment recommendations. We describe a treatment paradigm illustrated by a series of five patients who experienced irAEs. In three cases of metastatic melanoma, ipilimumab-induced hepatitis was successfully treated with high-dose parenteral pulsed methylprednisolone. In two other melanoma patients with ipilimumab-induced colitis, one patient had satisfactory resolution of his colitis with high-dose corticosteroid therapy alone and the other patient required infliximab infusion. We have reviewed the current literature and management algorithms for ipilimumab-induced irAEs. Treatment options and the rationale for their use are discussed, including the use of pulsed high-dose steroids, MMF, azathioprine and calcineurin inhibitors.
Collapse
Affiliation(s)
- Robert Cheng
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | | | | | | | | | | |
Collapse
|
306
|
Abstract
OBJECTIVES To present a thorough literature review on the assessment, grading, and treatment of rash associated with targeted therapies for cancer treatment. To identify ways that nursing can impact a patient's treatment experience by understanding and properly managing treatment for the rash. DATA SOURCES Peer-reviewed journal articles, textbooks. CONCLUSION Identification and management of rash induced by targeted therapies may improve quality of life and allow patients to continue drug therapy for their cancer to offer best outcomes. IMPLICATIONS FOR NURSING PRACTICE Nurses are in a unique position to assess, grade, and manage rash in patients receiving targeted therapies. Nurses will often be the first point of contact for the patient experiencing a rash, and the proper triage and advice on management can help the patient tolerate these drugs and enable them to remain on treatment.
Collapse
|
307
|
Mahzari M, Liu D, Arnaout A, Lochnan H. Immune checkpoint inhibitor therapy associated hypophysitis. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2015; 8:21-8. [PMID: 25861234 PMCID: PMC4376202 DOI: 10.4137/cmed.s22469] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/26/2015] [Accepted: 01/28/2015] [Indexed: 12/30/2022]
Abstract
Ipilimumab is a monoclonal antibody directed against CTLA4 T-lymphocyte antigen used as cancer therapy. Immune-related adverse events are common side effects and may include hypophysitis-related hypopituitarism. The clinical features of six patients with ipilimumab-induced hypophysitis (IH) are described. The clinical features of IH reported in clinical trials, including the incidence of IH by gender and the likelihood of adrenal axis recovery, are summarized. Following the development of IH, most patients remain on glucocorticoid replacement despite efforts to withdraw therapy. Analysis of gender information in published clinical trials suggests that men are more prone to developing IH than women, and few patients fully recover the pituitary–adrenal axis function. Ipilimumab and other drugs within its class are likely to be used to treat many forms of cancer. Endocrinologists should anticipate a significant increase in the incidence of autoimmune hypophysitis. Strategies for early detection of IH and long-term management should be considered.
Collapse
Affiliation(s)
- Moeber Mahzari
- Division of Endocrinology and Metabolism, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Dora Liu
- Division of Endocrinology and Metabolism, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Amel Arnaout
- Division of Endocrinology and Metabolism, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Heather Lochnan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| |
Collapse
|
308
|
Cytotoxic T-lymphocyte antigen-4 blockade in melanoma. Clin Ther 2015; 37:755-63. [PMID: 25746738 DOI: 10.1016/j.clinthera.2015.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 02/05/2023]
Abstract
PURPOSE Melanoma is an aggressive malignancy that has a complex relationship with the host immune system. Immunotherapies have long been a mainstay of melanoma therapy, and advanced therapies continue to be effective in treating this disease. Immune checkpoint blockade has proven to be a novel target in melanoma, with the approval of cytotoxic T-lymphocyte antigen-4 (CTLA-4)-targeted therapy. This review evaluates the role of CTLA-4-targeted therapies in the treatment of metastatic melanoma, with a focus on mechanisms, efficacy, toxicity, and future directions of this therapy. METHODS A search was performed in PubMed to identify relevant clinical studies that explored the clinical experience with CTLA-4-targeted therapy in melanoma. FINDINGS Signaling through CTLA-4 causes deactivation of T cells after the initial stimulatory signals. Therapies that block CTLA-4 lead to increased T-cell function and an antitumor response in patients with metastatic melanoma. The adverse event profile of these agents is different from that seen with more traditional cancer therapies and consists of dermatitis, colitis, and other autoimmune toxicities. In addition, the pattern of response is different from that seen with traditional cytotoxic therapies, with some patients experiencing initial progression followed by response and some patients having long-term durable responses. IMPLICATIONS Extensive clinical evidence supports the use of CTLA-4-targeted agents in the treatment of metastatic melanoma. The durability of response seen in patients receiving these agents has changed the landscape for patients with melanoma. Combination therapies and other agents with similar mechanisms warrant further exploration for the treatment of metastatic melanoma.
Collapse
|
309
|
Teulings HE, Limpens J, Jansen SN, Zwinderman AH, Reitsma JB, Spuls PI, Luiten RM. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol 2015; 33:773-81. [PMID: 25605840 DOI: 10.1200/jco.2014.57.4756] [Citation(s) in RCA: 436] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Vitiligo-like depigmentation in patients with melanoma may be associated with more favorable clinical outcome. We conducted a systematic review of patients with stage III to IV melanoma treated with immunotherapy to determine the cumulative incidence of vitiligo-like depigmentation and the prognostic value of vitiligo development on survival. METHODS We systemically searched and selected all studies on melanoma immunotherapy that reported on autoimmune toxicity and/or vitiligo between 1995 and 2013. Methodologic quality of each study was appraised using adapted criteria for systematic reviews in prognostic studies. Random-effect models were used to calculate summary estimates of the cumulative incidence of vitiligo-like depigmentation across studies. The prognostic value of vitiligo-like depigmentation on survival outcome was assessed using random-effects Cox regression survival analyses. RESULTS One hundred thirty-seven studies were identified comprising 139 treatment arms (11 general immune stimulation, 84 vaccine, 28 antibody-based, and 16 adoptive transfer) including a total of 5,737 patients. The overall cumulative incidence of vitiligo was 3.4% (95% CI, 2.5% to 4.5%). In 27 studies reporting individual patient data, vitiligo development was significantly associated with both progression-free-survival (hazard ratio [HR], 0.51; 95% CI, 0.32 to 0.82; P < .005) and overall survival (HR, 0.25; 95% CI, 0.10 to 0.61; P < .003), indicating that these patients have two to four times less risk of disease progression and death, respectively, compared with patients without vitiligo development. CONCLUSION Although vitiligo occurs only in a low percentage of patients with melanoma treated with immunotherapy, our findings suggest clear survival benefit in these patients. Awareness of vitiligo induction in patients with melanoma is important as an indicator of robust antimelanoma immunity and associated improved survival.
Collapse
Affiliation(s)
- Hansje-Eva Teulings
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Jacqueline Limpens
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sophia N Jansen
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aeilko H Zwinderman
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes B Reitsma
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Phyllis I Spuls
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Rosalie M Luiten
- Hansje-Eva Teulings, Jacqueline Limpens, Sophia N. Jansen, Aeilko H. Zwinderman, Johannes B. Reitsma, Phyllis I. Spuls, and Rosalie M. Luiten, Academic Medical Centre, University of Amsterdam, Amsterdam; Johannes B. Reitsma, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
310
|
Abstract
The anti-cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4) antibody ipilimumab is the first treatment that significantly improved the survival rates of metastatic melanoma patients, marking a new era in the treatment of melanoma. During its development, a hallmark of ipilimumab therapy was the extended duration of response, achieved in 20% of patients. The follow-up of patients included in phase II and phase III trials and in expanded access programs revealed that the survival rates remained stable after 3 years. These results demonstrated that ipilimumab induces an effective anti-tumor immune response persisting after the completion of treatment, and suggested a potential remission in a subset of patients. In this article we review the development of ipilimumab and highlight the long-term results. This approach emphasizes the need to optimize the use of ipilimumab in the future, by identifying the patients most likely to achieve long term survival after ipilimumab therapy, and by developing combined therapeutic approaches involving cytotoxic agents, targeted therapies or other immunotherapies to achieve durable control in a larger proportion of patients.
Collapse
Affiliation(s)
- Julie Delyon
- AP-HP, Hôpital Saint-Louis, Département de Dermatologie, Paris, France; INSERM U976, Paris 7 University, Paris, France
| | - Michele Maio
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | - Celeste Lebbé
- AP-HP, Hôpital Saint-Louis, Département de Dermatologie, Paris, France; INSERM U976, Paris 7 University, Paris, France; Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
311
|
Abstract
Since the development and approval of Ipilimumab, the first immune checkpoint inhibitor licensed for the treatment of metastatic melanoma, clinicians have gained a better understanding of the mode of action, management of toxicities, and assessment of response to this class of drugs. Several antibodies are now in development, aimed at blocking novel immune checkpoint molecules, such as PD-1 and it's corresponding ligand PD-L1. This article summarizes the mechanism of action, preclinical development, and subsequent clinical studies of immune checkpoint antibodies in melanoma.
Collapse
Affiliation(s)
- Jarushka Naidoo
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - David B Page
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jedd D Wolchok
- Melanoma and Immunotherapy Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA; Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
312
|
Abstract
Metastatic melanoma has an unpredictable natural history but a predictably high mortality. Despite recent advances in systemic therapy, many patients do not respond, or develop resistance to drug therapy. Surgery has consistently shown good outcomes in appropriately selected patients. It is likely to be even more successful in the era of more effective medical treatment. Surgery should remain a strongly considered option for metastatic melanoma.
Collapse
Affiliation(s)
- Gary B Deutsch
- Melanoma Research Program, John Wayne Cancer Institute, Providence St. John's Hospital, 2200 Santa Monica Boulevard, Santa Monica, CA 90404, USA
| | - Daniel D Kirchoff
- Melanoma Research Program, John Wayne Cancer Institute, Providence St. John's Hospital, 2200 Santa Monica Boulevard, Santa Monica, CA 90404, USA
| | - Mark B Faries
- Melanoma Research Program, John Wayne Cancer Institute, Providence St. John's Hospital, 2200 Santa Monica Boulevard, Santa Monica, CA 90404, USA.
| |
Collapse
|
313
|
Abstract
Immunologic checkpoint blockade with antibodies that target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death protein 1 pathway (PD-1/PD-L1) have demonstrated promise in a variety of malignancies. Ipilimumab (CTLA-4) and pembrolizumab (PD-1) are approved by the US Food and Drug Administration for the treatment of advanced melanoma, and additional regulatory approvals are expected across the oncologic spectrum for a variety of other agents that target these pathways. Treatment with both CTLA-4 and PD-1/PD-L1 blockade is associated with a unique pattern of adverse events called immune-related adverse events, and occasionally, unusual kinetics of tumor response are seen. Combination approaches involving CTLA-4 and PD-1/PD-L1 blockade are being investigated to determine whether they enhance the efficacy of either approach alone. Principles learned during the development of CTLA-4 and PD-1/PD-L1 approaches will likely be used as new immunologic checkpoint blocking antibodies begin clinical investigation.
Collapse
Affiliation(s)
- Michael A Postow
- All authors: Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Margaret K Callahan
- All authors: Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Jedd D Wolchok
- All authors: Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY.
| |
Collapse
|
314
|
Abstract
The aim of the study was to analyze the safety and efficacy of yttrium-90 ((90)Y) radioembolization in the treatment of unresectable hepatic melanoma metastases refractory to previous systemic/locoregional therapy. Between February 2004 and April 2010, 16 patients with hepatic melanoma metastases (ocular=7, skin=4, other sites=5) were treated with (90)Y radioembolization at a single center. Toxicity was assessed using the National Cancer Institute Common Terminology Criteria, version 3.0. Response to therapy was assessed by size and necrosis criteria. Progression-free survival (hepatic) and overall survival were calculated using the Kaplan-Meier method. The median dose to the treatment site was 108.57 Gy. Grade 1 and 2 clinical toxicities included fatigue (44%), nausea (19%), and vomiting (12%). Grade 3 absolute lymphocyte toxicity and aspartate aminotransferase toxicity were noted in 2 (12%) and 1 (7%) patients, respectively. Grade 4 bilirubin toxicity was observed in 1 (7%) patient. Overall, 13 (81%) patients showed disease control (response+stable disease) according to WHO, European Association for the Study of the Liver, and Response Evaluation Criteria for Solid Tumors. Progressive disease was observed in 3 (19%) patients according to WHO, European Association for the Study of the Liver, and Response Evaluation Criteria for Solid Tumors. The median overall and hepatic progression-free survival times were 7.63 and 4.23 months. Patients with disease control (responders+stable disease) survived longer than those with progressive disease (9.97 vs. 2.13 months, P<0.0001). Results from this small and single-center experience show that radioembolization is a safe therapy and its potential for being an efficacious therapy for hepatic melanoma metastases should be explored further. Radioembolization should be considered for liver-dominant disease refractory to other forms of systemic therapies.
Collapse
|
315
|
Min L, Hodi FS, Giobbie-Hurder A, Ott PA, Luke JJ, Donahue H, Davis M, Carroll RS, Kaiser UB. Systemic high-dose corticosteroid treatment does not improve the outcome of ipilimumab-related hypophysitis: a retrospective cohort study. Clin Cancer Res 2014; 21:749-55. [PMID: 25538262 DOI: 10.1158/1078-0432.ccr-14-2353] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE To examine the onset and outcome of ipilimumab-related hypophysitis and the response to treatment with systemic high-dose corticosteroids (HDS). EXPERIMENTAL DESIGN Twenty-five patients who developed ipilimumab-related hypophysitis were analyzed for the incidence, time to onset, time to resolution, frequency of resolution, and the effect of systemic HDS on clinical outcome. To calculate the incidence, the total number (187) of patients with metastatic melanoma treated with ipilimumab at Dana-Farber Cancer Institute (DFCI; Boston, MA) was retrieved from the DFCI oncology database. Comparisons between corticosteroid treatment groups were performed using the Fisher exact test. The distributions of overall survival were based on the method of Kaplan-Meier. RESULTS The overall incidence of ipilimumab-related hypophysitis was 13%, with a higher rate in males (16.1%) than females (8.7%). The median time to onset of hypophysitis after initiation of ipilimumab treatment was 9 weeks (range, 5-36 weeks). Resolution of pituitary enlargement, secondary adrenal insufficiency, secondary hypothyroidism, male secondary hypogonadism, and hyponatremia occurred in 73%, 0%, 64%, 45%, and 92% of patients, respectively. Systemic HDS treatment did not improve the outcome of hypophysitis as measured by resolution frequency and time to resolution. One-year overall survival in the cohort of patients was 83%, and while it was slightly higher in patients who did not receive HDS, there was no statistically significant difference between treatment arms. CONCLUSION Systemic HDS therapy in patients with ipilimumab-related hypophysitis may not be indicated. Instead, supportive treatment of hypophysitis-related hormone deficiencies with the corresponding hormone replacement should be given.
Collapse
Affiliation(s)
- Le Min
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Frank Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Jason J Luke
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Melanoma and Developmental Therapeutics Clinics, University of Chicago, Chicago, Illinois
| | - Hilary Donahue
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Meredith Davis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Rona S Carroll
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
316
|
Marlier J, Cocquyt V, Brochez L, Van Belle S, Kruse V. Ipilimumab, not just another anti-cancer therapy: hypophysitis as side effect illustrated by four case-reports. Endocrine 2014; 47:878-83. [PMID: 24554495 DOI: 10.1007/s12020-014-0199-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/31/2014] [Indexed: 12/14/2022]
Abstract
Ipilimumab is a monoclonal antibody that blocks cytotoxic T-lymphocyte antigen4 (CTLA-4), an inhibitory molecule typically expressed on T cells. Blockade of CTLA-4 induces an overall activation of T cells, including an immune-mediated anti-tumour response. Unfortunately, this broad T cell stimulation also causes immune-related adverse events (irAEs), such as dermatitis, colitis, hepatitis and hypophysitis. Ipilimumab is currently available in Belgium as a second line of treatment for patients with advanced melanoma, and is used at a dose of 3 mg/kg of body weight, although higher doses were previously used (up to 10 mg/kg). We performed a retrospective analysis to identify melanoma patients treated with ipilimumab at the Ghent University Hospital between 2010 and 2013. Data on symptoms, stage and timing of ipilimumab, response and adverse events were collected with a special attention to endocrine disturbances, going from a limited involvement of one endocrine axis to development of a hypophysitis. We identified a total of 39 patients with stage III (No. = 7) or stage IV (No. = 32) melanoma, who received a dose of 3 (No. = 31) or 10 (No. = 8) mg/kg. Six patients developed a severe form of irAEs, including one case of colitis (2 %), one case of sarcoidosis (2 %) and 4 cases (10 %) of hypophysitis. Hypophysitis developed between the second and fourth cycle of ipilimumab administration and was independent of the dose used. We describe four cases of involvement of the pituitary gland during treatment with ipilimumab. When managed with vigilant monitoring and high-dose corticosteroids, the acute symptoms resolve, but lifelong hormone substitution therapy can be necessary. Involvement of the pituitary axes is a severe side effect of treatment with ipilimumab with an urgent need for the correct medical intervention.
Collapse
Affiliation(s)
- Joke Marlier
- Department of Medical Oncology, University Hospital Ghent, De Pintelaan 185, 9000, Ghent, Belgium,
| | | | | | | | | |
Collapse
|
317
|
Page DB, Naidoo J, McArthur HL. Emerging immunotherapy strategies in breast cancer. Immunotherapy 2014; 6:195-209. [PMID: 24491092 DOI: 10.2217/imt.13.166] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Although immunogenicity is typically associated with renal cell carcinomas and melanoma, there are several compelling reasons why immune-based therapies should be explored in breast cancer. First, breast cancers express multiple putative tumor-associated antigens, such as HER-2 and MUC-1, which have been the successful focus of vaccine development over the past decade, translating into tumor-specific immune responses and, in some cases, clinical benefit. Second, passive immune strategies with anti-HER-2 antibodies, such as trastuzumab and pertuzumab, have led to survival benefits in breast cancer. Finally, the successes observed with novel immunotherapeutic strategies, such as immune checkpoint blockade and adoptive T-cell therapies in other malignancies, combined with a growing body of literature that supports an interplay between solid tumors and the immune system, indicate that these strategies have the potential to revolutionize the treatment of breast cancer.
Collapse
Affiliation(s)
- David B Page
- Memorial Sloan-Kettering Cancer Center, Department of Medicine, 300 East 66th Street, New York, NY 10065, NY, USA
| | | | | |
Collapse
|
318
|
Reardon DA, Freeman G, Wu C, Chiocca EA, Wucherpfennig KW, Wen PY, Fritsch EF, Curry WT, Sampson JH, Dranoff G. Immunotherapy advances for glioblastoma. Neuro Oncol 2014; 16:1441-58. [PMID: 25190673 DOI: 10.1093/neuonc/nou212] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Survival for patients with glioblastoma, the most common high-grade primary CNS tumor, remains poor despite multiple therapeutic interventions including intensifying cytotoxic therapy, targeting dysregulated cell signaling pathways, and blocking angiogenesis. Exciting, durable clinical benefits have recently been demonstrated for a number of other challenging cancers using a variety of immunotherapeutic approaches. Much modern research confirms that the CNS is immunoactive rather than immunoprivileged. Preliminary results of clinical studies demonstrate that varied vaccine strategies have achieved encouraging evidence of clinical benefit for glioblastoma patients, although multiple variables will likely require systematic investigation before optimal outcomes are realized. Initial preclinical studies have also revealed promising results with other immunotherapies including cell-based approaches and immune checkpoint blockade. Clinical studies to evaluate a wide array of immune therapies for malignant glioma patients are being rapidly developed. Important considerations going forward include optimizing response assessment and identifiying correlative biomarkers for predict therapeutic benefit. Finally, the potential of complementary combinatorial immunotherapeutic regimens is highly exciting and warrants expedited investigation.
Collapse
Affiliation(s)
- David A Reardon
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Gordon Freeman
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Catherine Wu
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - E Antonio Chiocca
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Kai W Wucherpfennig
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Edward F Fritsch
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - William T Curry
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - John H Sampson
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| | - Glenn Dranoff
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., P.Y.W.); Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Boston, Massachusetts (G.F., C.W., K.W.W.); Department of Medical Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (D.A.R., C.W.); Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts (E.A.C.); Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts (P.Y.W.); Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina (J.H.S.); Department of Neurosurgery, Massachusetts General Hospital, Boston, Massachusetts (W.T.C.); Department of Medical Oncology and Cancer Vaccine Center, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts (C.W., E.F.F., G.D.); Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts (G.D.)
| |
Collapse
|
319
|
Ishihara M, Seo N, Mitsui J, Muraoka D, Tanaka M, Mineno J, Ikeda H, Shiku H. Systemic CD8+ T cell-mediated tumoricidal effects by intratumoral treatment of oncolytic herpes simplex virus with the agonistic monoclonal antibody for murine glucocorticoid-induced tumor necrosis factor receptor. PLoS One 2014; 9:e104669. [PMID: 25105508 PMCID: PMC4126744 DOI: 10.1371/journal.pone.0104669] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/11/2014] [Indexed: 02/07/2023] Open
Abstract
Oncolytic virotherapy combined with immunomodulators is a novel noninvasive strategy for cancer treatment. In this study, we examined the tumoricidal effects of oncolytic HF10, a naturally occurring mutant of herpes simplex virus type-1, combined with an agonistic DTA-1 monoclonal antibody specific for the glucocorticoid-induced tumor necrosis factor receptor. Two murine tumor models were used to evaluate the therapeutic efficacies of HF10 virotherapy combined with DTA-1. The kinetics and immunological mechanisms of DTA-1 in HF10 infection were examined using flow cytometry and immunohistochemistry. Intratumoral administration of HF10 in combination with DTA-1 at a low dose resulted in a more vigorous attenuation of growth of the untreated contralateral as well as the treated tumors than treatment with either HF10 or DTA-1 alone. An accumulation of CD8+ T cells, including tumor- and herpes simplex virus type-1-specific populations, and a decrease in the number of CD4+ Foxp3+ T regulatory cells were seen in both HF10- and DTA-1-treated tumors. Studies using Fc-digested DTA-1 and Fcγ receptor knockout mice demonstrated the direct participation of DTA-1 in regulatory T cell depletion by antibody-dependent cellular cytotoxicity primarily via macrophages. These results indicated the potential therapeutic efficacy of a glucocorticoid-induced tumor necrosis factor receptor-specific monoclonal antibody in oncolytic virotherapy at local tumor sites.
Collapse
Affiliation(s)
- Mikiya Ishihara
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Naohiro Seo
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
- * E-mail: (NS); (HS)
| | - Jun Mitsui
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Daisuke Muraoka
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Maki Tanaka
- Gene Medicine Business Unit, Takara Bio Inc., Shiga, Japan
| | - Junichi Mineno
- Gene Medicine Business Unit, Takara Bio Inc., Shiga, Japan
| | - Hiroaki Ikeda
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
- * E-mail: (NS); (HS)
| |
Collapse
|
320
|
Ipilimumab treatment associated pituitary hypophysitis: clinical presentation and imaging diagnosis. Clin Neurol Neurosurg 2014; 125:125-30. [PMID: 25127260 DOI: 10.1016/j.clineuro.2014.06.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 06/05/2014] [Accepted: 06/08/2014] [Indexed: 01/18/2023]
Abstract
Ipilimumab is an immunomodulating drug for use in treatment of unresectable or metastatic melanoma with autoimmune lymphocytic hypophysitis as a reported complication. We describe three recent cases of ipilimumab associated autoimmune hypophysitis (IAH) at our institution, and provide a selected literature review showing its variable clinical presentation, imaging appearance and treatment in order to expedite early and appropriate IAH management. Patients had variable clinical presentation of hypophysitis, including headache, fatigue, visual changes, endocrinopathy, and/or hyponatremia. Contrast enhanced MRI showed symmetric pituitary gland and stalk enlargement in all of our cases and received a presumptive diagnosis of IAH. Following cessation of therapy and treatment there was normalization of pituitary morphology at follow-up MRI and return to clinical baseline. Varying clinical presentation can complicate the diagnosis of lymphocytic hypophysitis. One must be cognizant of its overall clinical and radiologic picture in patients receiving ipilimumab, now commonly used for the treatment of metastatic melanoma.
Collapse
|
321
|
Gilardi L, Grana CM, Paganelli G. Evaluation of response to immunotherapy: new challenges and opportunities for PET imaging. Eur J Nucl Med Mol Imaging 2014; 41:2090-2. [PMID: 25012872 DOI: 10.1007/s00259-014-2848-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 06/24/2014] [Indexed: 01/13/2023]
Affiliation(s)
- Laura Gilardi
- Division of Nuclear Medicine, European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy,
| | | | | |
Collapse
|
322
|
Simeone E, Grimaldi AM, Esposito A, Curvietto M, Palla M, Paone M, Mozzillo N, Ascierto PA. Serious haematological toxicity during and after ipilimumab treatment: a case series. J Med Case Rep 2014; 8:240. [PMID: 24986059 PMCID: PMC4090655 DOI: 10.1186/1752-1947-8-240] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/07/2014] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION Immunotherapy with the anti-cytotoxic T-lymphocyte antigen-4 monoclonal antibody ipilimumab has been shown to improve overall survival in previously treated and treatment-naïve patients with unresectable stage III or IV melanoma. Consistent with its proposed immunomodulating mechanism of action, the most common toxicities associated with ipilimumab therapy are immune-related in nature and include those related to the skin and gastrointestinal tract, with endocrine and hepatic events also frequent. Other rare adverse events, including haematological aberrations, may also occur and can have serious consequences if unrecognised. Here we describe three patients who developed serious haematological adverse events during or after treatment with ipilimumab. CASE PRESENTATION Three Caucasian patients (two women aged 68 and 49 years and one man aged 70 years) with metastatic melanoma experienced anaemia and/or leukopenia (neutropenia) with toxicity of various grades during or after treatment with ipilimumab, without significant changes to other haematological values. Two of the patients stopped treatment after the third ipilimumab dose, one because of severe anaemia that required blood transfusion and the other due to febrile neutropenia that was treated with antibiotics and granulocyte-macrophage colony-stimulating factor stimulation. The third patient developed anaemia and leukopenia after treatment during the follow-up period. The results of autoimmunity tests performed were positive and corticosteroids were used to treat these events as per side-effects treatment algorithms specifically developed for the management of immune-related adverse events associated with ipilimumab, an approach that was safe and effective. CONCLUSIONS Haematological toxicity is a rare but potentially serious immune-related side effect of ipilimumab therapy. However, if promptly recognised and treated, haematological toxicity is manageable and can be reversed with standard corticosteroid treatment as recommended for other ipilimumab immune-related side effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori Fondazione "G, Pascale", Via Mariano Semmola, 80131 Napoli, Italy.
| |
Collapse
|
323
|
Lacouture ME, Wolchok JD, Yosipovitch G, Kähler KC, Busam KJ, Hauschild A. Ipilimumab in patients with cancer and the management of dermatologic adverse events. J Am Acad Dermatol 2014; 71:161-9. [PMID: 24767731 DOI: 10.1016/j.jaad.2014.02.035] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 02/20/2014] [Accepted: 02/24/2014] [Indexed: 02/05/2023]
Abstract
Ipilimumab is a fully human monoclonal antibody that blocks cytotoxic T-lymphocyte antigen-4 to augment antitumor T-cell responses. Phase III studies have demonstrated survival benefit in both previously treated and treatment-naïve patients with metastatic melanoma. In clinical trials, adverse events (AEs) related to treatment with ipilimumab were mostly grade 1/2 (as per Common Terminology Criteria for AEs, Version 4.02), and mostly reversible with appropriate management. Distinct immune-related AEs that may reflect the mechanism of action of ipilimumab have been identified, and occur commonly in the skin, typically presenting as a maculopapular rash, which can be accompanied by pruritus, pruritus with no skin lesions, alopecia, and vitiligo. Histologic analyses have revealed epidermal spongiosis, and perivascular CD4(+) T-cell infiltrates with some eosinophils in areas of rash. Timely implementation of toxicity-specific treatment guidelines that emphasize vigilance and early intervention allows mitigation of dermatologic AEs. Adherence to guidelines is necessary to maintain quality of life, ensure consistent dosing, and obtain the best possible clinical outcome.
Collapse
Affiliation(s)
- Mario E Lacouture
- Memorial Sloan Kettering Cancer Center, New York, New York; Weill-Cornell Medical College, New York, New York.
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center, New York, New York; Weill-Cornell Medical College, New York, New York; Ludwig Institute for Cancer Research, New York, New York
| | - Gil Yosipovitch
- Departments of Dermatology, Neurobiology and Anatomy, and Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina
| | | | - Klaus J Busam
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Axel Hauschild
- Department of Dermatology, University of Kiel, Kiel, Germany
| |
Collapse
|
324
|
Chiarion Sileni V, Pigozzo J, Ascierto PA, Grimaldi AM, Maio M, Di Guardo L, Marchetti P, de Rosa F, Nuzzo C, Testori A, Cocorocchio E, Bernengo MG, Guida M, Marconcini R, Merelli B, Parmiani G, Rinaldi G, Aglietta M, Grosso M, Queirolo P. Efficacy and safety of ipilimumab in elderly patients with pretreated advanced melanoma treated at Italian centres through the expanded access programme. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:30. [PMID: 24708900 PMCID: PMC3996509 DOI: 10.1186/1756-9966-33-30] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/17/2014] [Indexed: 12/20/2022]
Abstract
Background Elderly patients with metastatic melanoma have different disease characteristics and a poorer prognosis than younger patients. Data from clinical trials and expanded access programmes (EAPs) suggest ipilimumab confers a consistent survival benefit and has a similar safety profile across different age groups of patients with metastatic melanoma. Here we report the efficacy and safety of ipilimumab 3 mg/kg in elderly patients enrolled in an EAP in Italy. Methods Patients aged > 70 years with pretreated melanoma received ipilimumab 3 mg/kg every 3 weeks for four doses through an EAP. Tumour response was evaluated at baseline and after completion of induction therapy using immune-related response criteria and patients were monitored throughout the treatment period for adverse events (AEs), including immune-related AEs. Results The immune-related disease control rate among 188 evaluable patients was 38%, including four patients with an immune-related complete response, 24 with an immune-related partial response and 44 with immune-related stable disease. Median progression-free survival (PFS) was 4.0 months and the 1- and 2-year PFS rates were 21% and 12%, respectively. Median overall survival (OS) was 8.9 months; 1- and 2-year OS rates were 38% and 22%, respectively. The safety profile of ipilimumab was consistent with that observed in the general population of the Italian EAP and treatment-related AEs generally resolved within a median of 2 weeks with treatment as per protocol-specific guidelines. Conclusions These results suggest ipilimumab is a feasible treatment option in elderly patients with metastatic melanoma. Ipilimumab treatment was generally well tolerated and resulted in clinical benefit and extended survival in elderly patients treated at centres in Italy.
Collapse
Affiliation(s)
- Vanna Chiarion Sileni
- Melanoma Cancer Unit, Oncology Institute of Veneto IRCCS, Via Gattamelata, 64, 35128 Padua, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
325
|
Luke JJ, Lezcano C, Hodi FS, Murphy GF. Antitumor granuloma formation by CD4+ T cells in a patient with rapidly progressive melanoma experiencing spiking fevers, neuropathy, and other immune-related toxicity after treatment with ipilimumab. J Clin Oncol 2014; 33:e32-5. [PMID: 24616309 DOI: 10.1200/jco.2013.49.7735] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Jason J Luke
- Melanoma Disease Center of Dana-Farber Cancer Institute; Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Cecilia Lezcano
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - F Stephen Hodi
- Melanoma Disease Center of Dana-Farber Cancer Institute; Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - George F Murphy
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
326
|
Ipilimumab before BRAF inhibitor treatment may be more beneficial than vice versa for the majority of patients with advanced melanoma. Cancer 2014; 120:1617-9. [DOI: 10.1002/cncr.28622] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/13/2013] [Indexed: 02/05/2023]
|
327
|
Jarkowski A, Khushalani NI. BRAF and beyond: Tailoring strategies for the individual melanoma patient. J Carcinog 2014; 13:1. [PMID: 24737949 PMCID: PMC3986540 DOI: 10.4103/1477-3163.126759] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 10/07/2013] [Indexed: 12/20/2022] Open
Abstract
Until recently, options for therapy in metastatic melanoma were limited. The understanding of immune check-point blockade and the discovery of molecular pathways involving driver mutations like BRAF has transformed the therapeutic landscape in this disease. Ipilimumab was the first drug shown to improve survival while vemurafenib demonstrated rapid responses never seen before in melanoma. Drugs from these classes and others are now in advanced stages of development and primed to positively impact patient survival in an incremental fashion. In this review, we highlight some of the developments during this renaissance in melanoma therapy and discuss agents of promise. Clinical challenges we face include individualizing therapy for patients, overcoming resistance to molecularly targeted therapy and developing rationale combinations or sequences of drugs. A concerted bench and bedside effort in this direction will undoubtedly keep melanoma in the forefront in an era of personalized medicine.
Collapse
Affiliation(s)
- Anthony Jarkowski
- Department of Pharmacy, James P. Wilmot Cancer Center, University of Rochester Medical Center, Rochester, USA
| | | |
Collapse
|
328
|
Bhatia S, Emdad L, Das SK, Hamed H, Dent P, Sarkar D, Fisher PB. Non-BRAF targeted therapies for melanoma: protein kinase inhibitors in Phase II clinical trials. Expert Opin Investig Drugs 2014; 23:489-500. [DOI: 10.1517/13543784.2014.884558] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
329
|
|
330
|
Barnes N, Chemaitilly W. Endocrinopathies in survivors of childhood neoplasia. Front Pediatr 2014; 2:101. [PMID: 25295241 PMCID: PMC4172013 DOI: 10.3389/fped.2014.00101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/06/2014] [Indexed: 01/26/2023] Open
Abstract
Advancements in cancer treatments have increased the number of survivors of childhood cancers. Endocrinopathies are common complications following cancer therapy and may occur decades later. The objective of the current review is to address the main endocrine abnormalities detected in childhood cancer survivors including disorders of the hypothalamic-pituitary axis, thyroid, puberty, gonads, bone, body composition, and glucose metabolism.
Collapse
Affiliation(s)
- Nicole Barnes
- Division of Pediatric Endocrinology, Department of Pediatric Medicine, St. Jude Children's Research Hospital , Memphis, TN , USA
| | - Wassim Chemaitilly
- Division of Pediatric Endocrinology, Department of Pediatric Medicine, St. Jude Children's Research Hospital , Memphis, TN , USA ; Epidemiology and Cancer Control, St. Jude Children's Research Hospital , Memphis, TN , USA
| |
Collapse
|
331
|
Quéreux G. [What’s new in dermato-oncology?]. Ann Dermatol Venereol 2013; 140 Suppl 3:S283-92. [PMID: 24365500 DOI: 10.1016/s0151-9638(13)70144-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
During this year 2013, Onco-dermatology was the object of numerous publications, especially in the field of metastatic melanoma. Previous results concerning anti PD-1 have been consolidated. Studies concerning Mek inhibitors have been published with promising results in uveal melanoma. In metastatic melanoma two combinations showed great results: combination of ipilimumab and nivolumab and combination of B-RAF and MEK inhibitors. Some studies demonstrated efficacy of these new therapeutics (ipilimumab, vemurafenib and dabrafenib) in brain metastasis. Moreover, the year 2013 was marked by the increasing knowledge in the management of adverse events induced by these new treatments. In the field of basal cell carcinoma, after the publication of large scale studies, vismodegib, the inhibitor of the hedgehog signalling pathway, was approved by the European Medicines Agency.
Collapse
Affiliation(s)
- G Quéreux
- Clinique dermatologique, CHU de Nantes, 1 place Alexis-Ricordeau, 44093 Nantes Cedex, France.
| |
Collapse
|
332
|
Silence K, Dreier T, Moshir M, Ulrichts P, Gabriels SME, Saunders M, Wajant H, Brouckaert P, Huyghe L, Van Hauwermeiren T, Thibault A, De Haard HJ. ARGX-110, a highly potent antibody targeting CD70, eliminates tumors via both enhanced ADCC and immune checkpoint blockade. MAbs 2013; 6:523-32. [PMID: 24492296 DOI: 10.4161/mabs.27398] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Overexpression of CD70 has been documented in a variety of solid and hematological tumors, where it is thought to play a role in tumor proliferation and evasion of immune surveillance. Here, we describe ARGX-110, a defucosylated IgG1 monoclonal antibody (mAb) that selectively targets and neutralizes CD70, the ligand of CD27. ARGX-110 was generated by immunization of outbred llamas. The antibody was germlined to 95% human identity, and its anti-tumor efficacy was tested in several in vitro assays. ARGX-110 binds CD70 with picomolar affinity. In depletion studies, ARGX-110 lyses tumor cells with greater efficacy than its fucosylated version. In addition, ARGX-110 demonstrates strong complement-dependent cytotoxicity and antibody-dependent cellular phagocytosis activity. ARGX-110 inhibits signaling of CD27, which results in blocking of the activation and proliferation of Tregs. In a Raji xenograft model, administration of the fucosylated version of ARGX-110 resulted in a prolonged survival at doses of 0.1 mg/kg and above. The pharmacokinetics of ARGX-110 was tested in cynomolgus monkeys; the calculated half-life is 12 days. In conclusion, ARGX-110 is a potent blocking mAb with a dual mode of action against both CD70-bearing tumor cells and CD70-dependent Tregs. This antibody is now in a Phase 1 study in patients with advanced malignancies expressing CD70 (NCT01813539).
Collapse
Affiliation(s)
| | | | | | | | | | | | - Harald Wajant
- Division of Molecular Internal Medicine; Department of Internal Medicine II; University Hospital Würzburg; Würzburg, Germany
| | - Peter Brouckaert
- VIB Department for Molecular Biomedical Research; Ghent University; Zwijnaarde, Belgium
| | - Leander Huyghe
- VIB Department for Molecular Biomedical Research; Ghent University; Zwijnaarde, Belgium
| | | | | | | |
Collapse
|
333
|
Torino F, Barnabei A, Paragliola RM, Marchetti P, Salvatori R, Corsello SM. Endocrine side-effects of anti-cancer drugs: mAbs and pituitary dysfunction: clinical evidence and pathogenic hypotheses. Eur J Endocrinol 2013; 169:R153-64. [PMID: 24001893 DOI: 10.1530/eje-13-0434] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
mAbs are established targeted therapies for several diseases, including hematological and solid malignancies. These agents have shown a favorable toxicity profile, but, despite their high selectivity, new typical side-effects have emerged. In cancer patients, pituitary dysfunction may be mainly due to brain metastases or primary tumors and to related surgery and radiotherapy. Anticancer agents may induce hypopituitarism in patients cured for childhood cancers. These agents infrequently affect pituitary function in adult cancer patients. Notably, hypophysitis, a previously very rare disease, has emerged as a distinctive side-effect of ipilimumab and tremelimumab, two mAbs inhibiting the cytotoxic T-lymphocyte antigen-4 receptor, being occasionally seen with nivolumab, another immune checkpoint inhibitor. Enhanced antitumor immunity is the suggested mechanism of action of these drugs and autoimmunity the presumptive mechanism of their toxicity. Recently, ipilimumab has been licensed for the treatment of patients affected by metastatic melanoma. With the expanding use of these drugs, hypophysitis will be progressively encountered by oncologists and endocrinologists in clinical practice. The optimal management of this potentially life-threatening adverse event needs a rapid and timely diagnostic and therapeutic intervention. Hypopituitarism caused by these agents is rarely reversible, requiring prolonged or lifelong substitutive hormonal treatment. Further studies are needed to clarify several clinical and pathogenic aspects of this new form of secondary pituitary dysfunction.
Collapse
Affiliation(s)
- Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | | | | | | | | | | |
Collapse
|
334
|
Page DB, Postow MA, Callahan MK, Allison JP, Wolchok JD. Immune modulation in cancer with antibodies. Annu Rev Med 2013; 65:185-202. [PMID: 24188664 DOI: 10.1146/annurev-med-092012-112807] [Citation(s) in RCA: 401] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ipilimumab is the prototypical immunomodulatory antibody, approved by the FDA in 2011 for advanced melanoma on the basis of survival benefit. Since that time, we have made significant strides in optimizing this therapy: we have characterized the spectrum of immune-related adverse events and learned how to mitigate them with treatment algorithms, discovered potential biomarkers of activity, and identified the potential synergy between checkpoint modulation and other therapeutic modalities. Recent phase I trials have established the efficacy and safety of next-generation checkpoint agents, including PD-1 and PD-L1 inhibitors, across multiple tumor types. Much work lies ahead in developing these next-generation checkpoint agents, testing them in combination, and determining how to integrate them into the treatment paradigms of various tumor types.
Collapse
Affiliation(s)
- David B Page
- Ludwig Center for Cancer Immunotherapy, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; , , ,
| | | | | | | | | |
Collapse
|
335
|
Min L, Hodi FS. Anti-PD1 following ipilimumab for mucosal melanoma: durable tumor response associated with severe hypothyroidism and rhabdomyolysis. Cancer Immunol Res 2013; 2:15-8. [PMID: 24778161 DOI: 10.1158/2326-6066.cir-13-0146] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Treatment with fully human monoclonal antibodies against programmed death 1 (PD1) receptor has shown great promise for a number of advanced malignancies. Although inflammatory adverse events have been well described with anti-CTL antigen 4 (CTLA4) therapy, experience with the range of adverse effects of anti-PD1 remains comparatively limited. Here, we report on a patient with advanced mucosal melanoma who received four doses of MK-3475, a fully human monoclonal antibody against PD1, and experienced a durable near-complete response but developed severe hypothyroidism, rhabdomyolysis, and acute kidney injury. To our knowledge, this is the first case reported of a patient with advanced mucosal melanoma who responded to anti-PD1 therapy. With the promising antitumor effects of anti-PD1 in a wide array of tumors, we expect an increasing number of patients to be exposed to anti-PD1 therapies. Recognition of infrequent presentations of adverse events such as elevated creatine kinase levels and thyroid disorders in patients who receive anti-PD1 therapy is important.
Collapse
Affiliation(s)
- Le Min
- Authors' Affiliations: Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | |
Collapse
|
336
|
Audemard A, de Raucourt S, Miocque S, Comoz F, Giraud JM, Dreno B, Bienvenu B, Rogerie MJ, Dompmartin A. Melanoma-associated retinopathy treated with ipilimumab therapy. Dermatology 2013; 227:146-9. [PMID: 24051549 DOI: 10.1159/000353408] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 06/02/2013] [Indexed: 11/19/2022] Open
Abstract
Melanoma-associated retinopathy (MAR) is a rare autoimmune syndrome in patients with melanoma characterized by visual disorders. MAR is induced by the degeneration of bipolar cells of the retina and the presence of serum autoantibodies against retina proteins. Ipilimumab, an anti-cytotoxic T lymphocyte-associated antigen 4 antibody, improves survival in previously treated patients with metastatic melanoma, but is responsible for a spectrum of immune-related adverse events. Administration of ipilimumab to patients with autoimmune diseases (such as MAR or vitiligo) is actually not recommended. We report a patient presenting with MAR occurring during a melanoma relapse. Surgery and chemotherapy had no effect on visual acuity and melanoma increased. In the absence of alternative antitumoral treatment, we focused on the vital prognosis and treated the patient with ipilimumab. Two years after the treatment the patient is free from new metastasis but has presented with exacerbation of vitiligo and MAR. In the very rare case of melanoma with autoimmune disease without a therapy option, ipilimumab could be discussed, taking into account the fact that it can be effective on tumor burden but can also increase autoimmunity.
Collapse
Affiliation(s)
- A Audemard
- Department of Internal Medicine, Caen, France
| | | | | | | | | | | | | | | | | |
Collapse
|
337
|
Müller D. Antibody–Cytokine Fusion Proteins for Cancer Immunotherapy: An Update on Recent Developments. BioDrugs 2013; 28:123-31. [DOI: 10.1007/s40259-013-0069-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|