151
|
Borzillo V, Di Franco R, Giannarelli D, Cammarota F, Scipilliti E, D’Ippolito E, Petito A, Serra M, Falivene S, Grimaldi AM, Simeone E, Festino L, Vanella V, Trojaniello C, Vitale MG, Madonna G, Ascierto PA, Muto P. Ipilimumab and Stereotactic Radiosurgery with CyberKnife ® System in Melanoma Brain Metastases: A Retrospective Monoinstitutional Experience. Cancers (Basel) 2021; 13:cancers13081857. [PMID: 33924595 PMCID: PMC8068853 DOI: 10.3390/cancers13081857] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 03/27/2021] [Accepted: 04/07/2021] [Indexed: 01/22/2023] Open
Abstract
Simple Summary Retrospective studies have shown a survival advantage in combining ipilimumab with radiotherapy in patients with melanoma brain metastases (MBMs). However, these studies did not clarify the correct timing between the two methods. The aims of our study were to demonstrate the efficacy and toxicity of stereotactic radiotherapy/radiosurgery on MBMs in combination with ipilimumab and estimate the correct timing of treatments to improve patients’ outcomes. Abstract The median overall survival (OS) and local control (LC) of patients with melanoma brain metastases (MBMs) are poor even with immune checkpoint inhibitors and/or radiotherapy (RT). The aims of the study were to evaluate the association and timing of stereotactic radiotherapy (SRT)/radiosurgery (SRS) performed with the CyberKnife® System and ipilimumab (IPI). A total of 63 MBMs patients were analyzed: 53 received RT+IPI and 10 RT alone. Therefore, the patients were divided into four groups: RT PRE-PI (>4 weeks before IPI) (18), RT CONC-IPI (4 weeks before/between first and last cycle/within 3 months of last cycle of IPI) (20), RT POST-IPI (>3 months after IPI) (15), and NO-IPI (10). A total of 127 lesions were treated: 75 with SRS (one fraction) and 24 with SRT (three to five fractions). The median follow-up was 10.6 months. The median OS was 10.6 months for all patients, 10.7 months for RT+IPI, and 3.3 months for NO-IPI (p = 0.96). One-year LC was 50% for all patients, 56% for RT+IPI, and 18% for NO-IPI (p = 0.08). The 1-year intracranial control was 45% for all patients, 44% for RT+IPI, and 51% for NO-IPI (p = 0.73). IPI with SRS/SRT in MBMs treatment could improve LC. However, the impact and timing of the two modalities on patients’ outcomes are still unclear.
Collapse
Affiliation(s)
- Valentina Borzillo
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
- Correspondence: ; Tel.: +39-08159031764; Fax: +39-0815903809
| | - Rossella Di Franco
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| | - Diana Giannarelli
- Statistical Unit, Regina Elena National Cancer Institute-IRCCS, 00144 Rome, Italy;
| | - Fabrizio Cammarota
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| | - Esmeralda Scipilliti
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| | - Emma D’Ippolito
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| | - Angela Petito
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| | - Marcello Serra
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| | - Sara Falivene
- Radiation Oncology Unit, Ospedale del Mare, Asl Napoli 1 Centro, 80147 Naples, Italy;
| | - Antonio M. Grimaldi
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Ester Simeone
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Lucia Festino
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Claudia Trojaniello
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Maria Grazia Vitale
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Paolo A. Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.M.G.); (E.S.); (L.F.); (V.V.); (C.T.); (M.G.V.); (G.M.); (P.A.A.)
| | - Paolo Muto
- Radiation Oncology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.D.F.); (F.C.); (E.S.); (E.D.); (A.P.); (M.S.); (P.M.)
| |
Collapse
|
152
|
Neoadjuvant Cytoreductive Treatment With BRAF/MEK Inhibition of Prior Unresectable Regionally Advanced Melanoma to Allow Complete Surgical Resection, REDUCTOR: A Prospective, Single-arm, Open-label Phase II Trial. Ann Surg 2021; 274:383-389. [PMID: 33843797 DOI: 10.1097/sla.0000000000004893] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To evaluate the potency of short-term neoadjuvant cytoreductive therapy with dabrafenib plus trametinib (BRAF and MEK inhibitor) to allow for radical surgical resection in patients with unresectable locally advanced melanoma. SUMMARY BACKGROUND DATA Approximately 5% of stage III melanoma patients presents with unresectable locally advanced disease, making standard of care with resection followed by adjuvant systemic therapy impossible. Although neoadjuvant targeted therapy has shown promising results in resectable stage III melanoma, its potency to enable surgical resection in patients with primarily unresectable locally advanced stage III melanoma is still unclear. METHODS In this prospective, single-arm, phase II trial, patients with unresectable BRAF-mutated locally advanced stage IIIC or oligometastatic stage IV melanoma were included. After 8 weeks of treatment with dabrafenib and trametinib, evaluation by positron emission tomography/computed tomography and physical examination were used to assess sufficient downsizing of the tumor to enable resection. The primary objective was the percentage of patients who achieved a radical (R0) resection. RESULTS Between August 2014 and March 2019, 21 patients (20/21 stage IIIC American Joint Committee on Cancer staging manual 7th edition) were included. Planned inclusion of 25 patients was not reached due to slow accrual and changing treatment landscape. Despite this, the predefined endpoint was successfully met. In 18/21 (86%) patients a resection was performed, of which 17 were R0 resections. At a median follow-up of 50 months (interquartile range 37.7-57.1 months), median recurrence-free survival was 9.9 months (95% confidence interval 7.52-not reached) in patients undergoing surgery. CONCLUSIONS This prospective, single-arm, open-label phase II trial, shows neoadjuvant dabrafenib plus trametinib as a potent cytoreductive treatment, allowing radical resection of metastases in 17/21 (81%) patients with prior unresectable locally advanced melanoma.
Collapse
|
153
|
Yap TA, Parkes EE, Peng W, Moyers JT, Curran MA, Tawbi HA. Development of Immunotherapy Combination Strategies in Cancer. Cancer Discov 2021; 11:1368-1397. [PMID: 33811048 DOI: 10.1158/2159-8290.cd-20-1209] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 01/03/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022]
Abstract
Harnessing the immune system to treat cancer through inhibitors of CTLA4 and PD-L1 has revolutionized the landscape of cancer. Rational combination strategies aim to enhance the antitumor effects of immunotherapies, but require a deep understanding of the mechanistic underpinnings of the immune system and robust preclinical and clinical drug development strategies. We review the current approved immunotherapy combinations, before discussing promising combinatorial approaches in clinical trials and detailing innovative preclinical model systems being used to develop rational combinations. We also discuss the promise of high-order immunotherapy combinations, as well as novel biomarker and combinatorial trial strategies. SIGNIFICANCE: Although immune-checkpoint inhibitors are approved as dual checkpoint strategies, and in combination with cytotoxic chemotherapy and angiogenesis inhibitors for multiple cancers, patient benefit remains limited. Innovative approaches are required to guide the development of novel immunotherapy combinations, ranging from improvements in preclinical tumor model systems to biomarker-driven trial strategies.
Collapse
Affiliation(s)
- Timothy A Yap
- Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eileen E Parkes
- Oxford Institute of Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Justin T Moyers
- Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
154
|
Goyal I, Pandey MR, Sharma R, Chaudhuri A, Dandona P. The side effects of immune checkpoint inhibitor therapy on the endocrine system. Indian J Med Res 2021; 154:559-570. [PMID: 35435341 PMCID: PMC9205006 DOI: 10.4103/ijmr.ijmr_313_19] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a relatively newer class of drugs approved for the treatment of malignancies such as melanoma, renal, bladder and lung cancer. Immune-related adverse events (IrAEs) involving the endocrine system are a common side effect of these drugs. The spectrum of endocrine adverse events varies by the drug class. Cytotoxic T-lymphocyte–associated antigen-4 inhibitors commonly cause hypophysitis/hypopituitarism, whereas the incidence of thyroid disease is higher with programmed cell death (PD)-1/ ligand (PD-L) protein 1 inhibitors. The focus of this review is to describe the individual endocrinopathies with their possible mechanisms, signs and symptoms, clinical assessment and disease management. Multiple mechanisms of IrAEs have been described in literature including type II/IV hypersensitivity reactions and development of autoantibodies. Patients with pre-existing autoimmune endocrine diseases can have disease exacerbation following ICI therapy rather than de novo IrAEs. Most of the endocrinopathies are relatively mild, and timely hormone replacement therapy allows continuation of ICIs. However, involvement of the pituitary–adrenal axis could be life-threatening if not recognized. Corticosteroids are helpful when the pituitary–adrenal axis is involved. In cases of severe endocrine toxicity (grade 3/4), ICIs should be temporarily discontinued and can be restarted after adequate hormonal therapy. Endocrinologists and general internists need to be vigilant and maintain a high degree of awareness for these adverse events.
Collapse
Affiliation(s)
- Itivrita Goyal
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo, Buffalo, NY, USA
| | - Manu Raj Pandey
- Department of Hematology & Oncology, State University of New York at Buffalo; Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Rajeev Sharma
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo; Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ajay Chaudhuri
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo, Buffalo, NY, USA
| | - Paresh Dandona
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
155
|
George AS, Fernandez CJ, Eapen D, Pappachan JM. Organ-specific Adverse Events of Immune Checkpoint Inhibitor Therapy, with Special Reference to Endocrinopathies. TOUCHREVIEWS IN ENDOCRINOLOGY 2021; 17:21-32. [PMID: 35118443 PMCID: PMC8320015 DOI: 10.17925/ee.2021.17.1.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/09/2021] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitors are potent and promising immunotherapeutic agents that are increasingly used for the management of various types of advanced cancers. The widespread approval of this group of drugs simultaneously revealed immune-related adverse events as unique side-effects. Endocrinopathies are one of the most common immune-related adverse events. The precise pathogenic mechanisms for these endocrinopathies are still unclear. Though few of the endocrinopathies are reversible, calling for only symptom control, most are irreversible, requiring multiple long-term hormone replacement therapies. However, in contrast to other organ-specific immune-related adverse events, patients with endocrinopathies can continue their immune checkpoint therapy, provided the hormone replacement therapy is adequate and the symptoms are controlled. Though patients who have developed immune-related adverse events demonstrate superior antitumor activity and overall survival, due to the high morbidity associated with the immune-related adverse events, researchers are trying to uncouple the antitumour activity associated with immune checkpoint inhibitor therapy from the immune-related adverse events, to preserve antitumour activity without adverse events.
Collapse
Affiliation(s)
- Annu Susan George
- Department of Medical Oncology, Lakeshore Hospital, Cochin, Kerala, India
| | - Cornelius J Fernandez
- Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK
| | - Dilip Eapen
- Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK
| | - Joseph M Pappachan
- Department of Endocrinology & Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston, UK
- Manchester Metropolitan University, Manchester, UK
- The University of Manchester, Manchester, UK
| |
Collapse
|
156
|
Deligiorgi MV, Liapi C, Trafalis DT. Hypophysitis related to immune checkpoint inhibitors: An intriguing adverse event with many faces. Expert Opin Biol Ther 2021; 21:1097-1120. [PMID: 33393372 DOI: 10.1080/14712598.2021.1869211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The incorporation of immune checkpoint inhibitors in the oncologists' arsenal is a milestone in cancer therapeutics, though not being devoid of toxicities.Areas covered: The present review provides a comprehensive and up-to-date overview of the immune-related hypophysitis with focus on the elusive biological background, the wide spectrum of the epidemiological profile, the varying clinical aspects, and the diagnostic and therapeutic challenges.Expert opinion: Historically considered distinctive of anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) monoclonal antibodies (mAbs), the immune-related hypophysitis is increasingly correlated with the anti-programmed cell-death (PD) protein 1 (PD-1)/anti-PD ligand 1 (PD-L1) mAbs. The distinct phenotype of hypophysitis related to anti-PD1/anti-PD-L1 mAbs is highlighted with focus on the immune-related isolated adrenocorticotropic (ACTH) deficiency. The immune-related central diabetes insipidus is discussed as a rare aspect of anti-CTL-A4 mAbs-induced hypophysitis, recently related to anti-PD1/anti-PD-L1 mAbs as well. The present review builds on existing literature concerning immune-related hypophysitis underscoring the pending issues still to be addressed, including (i) pathogenesis; (ii) correlation with preexisting autoimmunity; (iii) predictive value; (iv) utility of high-dose glucocorticoids; and (v) establishment of evidence-based diagnostic and therapeutic protocols. Increased awareness and constant vigilance are advocated as cornerstone of a multidisciplinary approach to ensure optimal patients' care.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology-Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| | - Charis Liapi
- Department of Pharmacology-Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology-Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine, Athens, Greece
| |
Collapse
|
157
|
Olofsson Bagge R, Ny L, Ascierto PA, Hodi FS, Larkin J, Robert C, Schachter J, Weber JS, Long GV, van Akkooi ACJ. The efficacy of immunotherapy for in-transit metastases of melanoma: an analysis of randomized controlled trials. Melanoma Res 2021; 31:181-185. [PMID: 33625104 DOI: 10.1097/cmr.0000000000000719] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nearly 10% of patients with high-risk early-stage melanoma will develop satellite or in-transit metastases (ITM), classified as stage III disease similar to lymph node metastases. The pivotal registration trials of the CTLA-4 antibody ipilimumab, and the PD-1 antibodies nivolumab and pembrolizumab, also included patients with unresectable stage III disease. However, there has been no analysis of patients with ITM, and anecdotal retrospective small series have indicated a potential lesser effect. This study aimed to identify patients with unresectable ITM within the randomized trials, and to determine response, progression-free survival and overall survival. The pivotal phase III randomized intervention trials that included melanoma patients with ITM, with or without nodal metastasis, and were treated with ipilimumab, nivolumab or pembrolizumab was identified. The datasets from each trial were then searched to identify the specific details of the investigated patient population for a pooled analysis. The primary endpoint was complete response rate. Seven trials that included stage III patients, and with accessible datasets, were identified. There was a total of 4711 patients, however, no patients with ITM could be identified, as this data was not captured by the case report forms. Evidence from prospective clinical trials on the use of immunotherapy in patients with ITM is lacking. We recommend pooling data from multiple institutions to examine efficacy of available drug therapies in this patient population, but more importantly, prospective clinical trials of locoregional treatments with or without systemic drug therapies are required.
Collapse
Affiliation(s)
- Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg
| | - Lars Ny
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Paolo A Ascierto
- Department of Medical Oncology, Istituto Nazionale Tumori IRCCS Fondazione 'G. Pascale', Naples, Italy
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - James Larkin
- Department of Medical Oncology, The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Caroline Robert
- Department of Medical Oncology Gustave Roussy and Paris-Saclay University, Villejuif, France
| | - Jacob Schachter
- Department of Medical Oncology, Ella Lemelbaum Institute for Immuno Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Jeffrey S Weber
- Department of Medical Oncology, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York City, New York, USA
| | - Georgina V Long
- Department of Medical Oncology, Melanoma Institute Australia, The University of Sydney and Royal North Shore and Mater Hospitals, Sydney, Australia
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
158
|
Abstract
The management of melanoma significantly improved within the last 25 years. Chemotherapy was the first approved systemic therapeutic approach and resulted in a median overall of survival less than 1 year, without survival improvement in phase III trials. High-dose interferon α2b and IL-2 were introduced for resectable high-risk and advanced disease, respectively, resulting in improved survival and response rates. The anti-CTLA4 and anti-programmed death 1 monoclonal antibodies along with BRAF/MEK targeted therapies are the dominant therapeutic classes of agent for melanoma. This article provides an historic overview of the evolution of melanoma management.
Collapse
|
159
|
Shimozaki K, Hirata K, Horie S, Chida A, Tsugaru K, Hayashi Y, Kawasaki K, Miyanaga R, Hayashi H, Mizuno R, Funakoshi T, Hosoe N, Hamamoto Y, Kanai T. The Entire Intestinal Tract Surveillance Using Capsule Endoscopy after Immune Checkpoint Inhibitor Administration: A Prospective Observational Study. Diagnostics (Basel) 2021; 11:543. [PMID: 33803735 PMCID: PMC8003297 DOI: 10.3390/diagnostics11030543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite the proven efficacy of immune checkpoint inhibitors (ICIs) against various types of malignancies, they have been found to induce immune-related adverse events, such as enterocolitis; however, the clinical features of ICI-induced enterocolitis remain to be sufficiently elucidated, which is significant, considering the importance of early detection in the appropriate management and treatment of ICI-induced enterocolitis. Therefore, the current study aimed to determine the utility of capsule endoscopy as a screening tool for ICI-induced enterocolitis. METHODS This single-center, prospective, observational study was conducted on patients with malignancy who received any ICI between April 2016 and July 2020 at Keio University Hospital. Next, second-generation capsule endoscopy (CCE-2) was performed on day 60 after ICI initiation to explore the entire gastrointestinal tract. RESULTS Among the 30 patients enrolled herein, 23 underwent CCE-2. Accordingly, a total of 23 findings were observed in 14 (60.8%) patients at any portion of the gastrointestinal tract (7 patients in the colon, 4 patients in the small intestine, 2 patients in both the colon and the small intestine, and 1 patient in the stomach). After capsule endoscopy, 2 patients (8.7%) developed ICI-induced enterocolitis: both had significantly higher Capsule Scoring of Ulcerative Colitis than those who had not developed ICI-induced enterocolitis (p = 0.0455). No adverse events related to CCE-2 were observed. CONCLUSIONS CCE-2 might be a safe and useful entire intestinal tract screening method for the early detection of ICI-induced enterocolitis in patients with malignancies.
Collapse
Affiliation(s)
- Keitaro Shimozaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Sara Horie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Akihiko Chida
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Kai Tsugaru
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Yukie Hayashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Kenta Kawasaki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Ryoichi Miyanaga
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| | - Hideyuki Hayashi
- Keio Cancer Center, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.H.); (Y.H.)
| | - Ryuichi Mizuno
- Department of Urology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Naoki Hosoe
- Center for Diagnostic and Therapeutic Endoscopy, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Yasuo Hamamoto
- Keio Cancer Center, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.H.); (Y.H.)
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan; (K.S.); (S.H.); (A.C.); (K.T.); (Y.H.); (K.K.); (R.M.); (T.K.)
| |
Collapse
|
160
|
Karimi A, Alilou S, Mirzaei HR. Adverse Events Following Administration of Anti-CTLA4 Antibody Ipilimumab. Front Oncol 2021; 11:624780. [PMID: 33767992 PMCID: PMC7985548 DOI: 10.3389/fonc.2021.624780] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 02/02/2021] [Indexed: 12/19/2022] Open
Abstract
Ipilimumab, a monoclonal anti-CTLA4 antibody, paved the path for promising treatments, particularly in advanced forms of numerous cancers like melanoma. By blockading CTLA-4, ipilimumab can abolish the higher binding affinity of B7 for CTLA-4, setting CD28 free to act unlimited. This blockade can result in an amplified antitumor immune response, and thereby, boosting more effective tumor regression. However, this blockage can lead to diminished self-tolerance and yielding autoimmune complications. The current review aims to describe adverse events (AEs) following the administration of ipilimumab in different cancers as every benefit comes at a cost. We will also discuss AEs in two different categories, melanoma and non-melanoma, owing to the possible shining promises in treating non-melanoma cancers. As the melanoma settings are more studied than other cancers, it might even help predict the patterns related to the other types of cancers. This similarity also might help physicians to predict adverse events and correctly manage them in non-melanoma cancers using the extensive findings reported in the more-studied melanoma settings. Recognizing the adverse events is vital since most of the adverse events could be reverted while carefully implementing guidelines. Finally, we will also describe the observed effectiveness of ipilimumab in non-melanoma cancers. This effectiveness reveals the importance of understanding the profile of adverse events in this group, even though some have not received FDA approval yet. Further clinical trials and careful systematic reviews may be required to decipher the hidden aspects of therapies with ipilimumab and its related AEs.
Collapse
Affiliation(s)
- Amirali Karimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanam Alilou
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
161
|
Cremolini C, Vitale E, Rastaldo R, Giachino C. Advanced Nanotechnology for Enhancing Immune Checkpoint Blockade Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:661. [PMID: 33800368 PMCID: PMC7998763 DOI: 10.3390/nano11030661] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/28/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022]
Abstract
Immune checkpoint receptor signaling pathways constitute a prominent class of "immune synapse," a cell-to-cell connection that represses T-lymphocyte effector functions. As a possible evolutionary countermeasure against autoimmunity, this strategy is aimed at lowering potential injury to uninfected cells in infected tissues and at minimizing systemic inflammation. Nevertheless, tumors can make use of these strategies to escape immune recognition, and consequently, such mechanisms represent chances for immunotherapy intervention. Recent years have witnessed the advance of pharmaceutical nanotechnology, or nanomedicine, as a possible strategy to ameliorate immunotherapy technical weaknesses thanks to its intrinsic biophysical properties and multifunctional modifying capability. To improve the long-lasting response rate of checkpoint blockade therapy, nanotechnology has been employed at first for the delivery of single checkpoint inhibitors. Further, while therapy via single immune checkpoint blockade determines resistance and a restricted period of response, strong interest has been raised to efficiently deliver immunomodulators targeting different inhibitory pathways or both inhibitory and costimulatory pathways. In this review, the partially explored promise in implementation of nanotechnology to improve the success of immune checkpoint therapy and solve the limitations of single immune checkpoint inhibitors is debated. We first present the fundamental elements of the immune checkpoint pathways and then outline recent promising results of immune checkpoint blockade therapy in combination with nanotechnology delivery systems.
Collapse
Affiliation(s)
- Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Emanuela Vitale
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy; (E.V.); (C.G.)
| | - Raffaella Rastaldo
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy; (E.V.); (C.G.)
| | - Claudia Giachino
- Department of Clinical and Biological Sciences, University of Torino, 10043 Orbassano, Italy; (E.V.); (C.G.)
| |
Collapse
|
162
|
Majem M, Manzano JL, Marquez-Rodas I, Mujika K, Muñoz-Couselo E, Pérez-Ruiz E, de la Cruz-Merino L, Espinosa E, Gonzalez-Cao M, Berrocal A. SEOM clinical guideline for the management of cutaneous melanoma (2020). Clin Transl Oncol 2021; 23:948-960. [PMID: 33651321 PMCID: PMC8057998 DOI: 10.1007/s12094-020-02539-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
Melanoma affects about 6000 patients a year in Spain. A group of medical oncologists from Spanish Society of Medical Oncology (SEOM) and Spanish Multidisciplinary Melanoma Group (GEM) has designed these guidelines to homogenize the management of these patients. The diagnosis must be histological and determination of BRAF status has to be performed in patients with stage ≥ III. Stage I–III resectable melanomas will be treated surgically. In patients with stage III melanoma, adjuvant treatment with immunotherapy or targeted therapy is also recommended. Patients with unresectable or metastatic melanoma will receive treatment with immunotherapy or targeted therapy, the optimal sequence of these treatments remains unclear. Brain metastases require a separate consideration, since, in addition to systemic treatment, they may require local treatment. Patients must be followed up closely to receive or change treatment as soon as their previous clinical condition changes, since multiple therapeutic options are available.
Collapse
Affiliation(s)
- M Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, c/Sant Antoni Maria Claret 167, 08025, Barcelona, Spain.
| | - J L Manzano
- Department of Medical Oncology, H. Germans Trias i Pujol, Catalan Institute of Oncology, ICO-Badalona, Badalona, Spain
| | - I Marquez-Rodas
- Department of Medical Oncology, Instituto de Investigación Sanitaria Gregorio Marañón and CIBERONC, Madrid, Spain
| | - K Mujika
- Department of Medical Oncology, UGC de Oncología de Gipuzkoa, OSI Donostialdea-Onkologikoa, Guipúzcoa, Spain
| | - E Muñoz-Couselo
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), Hospital Vall d'Hebron Barcelona, Barcelona, Spain
| | - E Pérez-Ruiz
- Department of Medical Oncology, Hospital Costa del Sol and UGC Oncol, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional Virgen Victoria, Málaga, Spain
| | - L de la Cruz-Merino
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain.,Medicine Department, Universidad de Sevilla, Seville, Spain
| | - E Espinosa
- Department of Medical Oncology, Hospital Universitario La Paz, CIBERONC, Madrid, Spain
| | - M Gonzalez-Cao
- Oncology Department (IOR), Hospital Dexeus, Barcelona, Spain
| | - A Berrocal
- Department of Medical Oncology, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| |
Collapse
|
163
|
Vreeland TJ, Clifton GT, Hale DF, Chick RC, Hickerson AT, Cindass JL, Adams AM, Bohan PMK, Andtbacka RHI, Berger AC, Jakub JW, Sussman JJ, Terando AM, Wagner T, Peoples GE, Faries MB. A Phase IIb Randomized Controlled Trial of the TLPLDC Vaccine as Adjuvant Therapy After Surgical Resection of Stage III/IV Melanoma: A Primary Analysis. Ann Surg Oncol 2021; 28:6126-6137. [PMID: 33641012 PMCID: PMC7914039 DOI: 10.1245/s10434-021-09709-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/25/2021] [Indexed: 12/15/2022]
Abstract
Background Melanoma therapy has changed dramatically over the last decade with improvements in immunotherapy, yet many patients do not respond to current therapies. This novel vaccine strategy may prime a patient’s immune system against their tumor and work synergistically with immunotherapy against advanced-stage melanoma. Methods This was a prospective, randomized, double-blind, placebo-controlled, phase IIb trial of the tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine administered to prevent recurrence in patients with resected stage III/IV melanoma. Patients were enrolled and randomized 2:1 to the TLPLDC vaccine or placebo (empty yeast cell wall particles and autologous dendritic cells). Both intention-to-treat (ITT) and per treatment (PT) analyses were predefined, with PT analysis including patients who remained disease-free through the primary vaccine/placebo series (6 months). Results A total of 144 patients were randomized (103 vaccine, 41 control). Therapy was well-tolerated with similar toxicity between treatment arms; one patient in each group experienced related serious adverse events. While disease-free survival (DFS) was not different between groups in ITT analysis, in PT analysis the vaccine group showed improved 24-month DFS (62.9% vs. 34.8%, p = 0.041). Conclusions This phase IIb trial of TLPLDC vaccine administered to patients with resected stage III/IV melanoma shows TLPLDC is well-tolerated and improves DFS in patients who complete the primary vaccine series. This suggests patients who do not recur early benefit from TLPLDC in preventing future recurrence from melanoma. A phase III trial of TLPLDC + checkpoint inhibitor versus checkpoint inhibitor alone in patients with advanced, surgically resected melanoma is under development. Trial Registration NCT02301611. Supplementary information The online version contains supplementary material available at (10.1245/s10434-021-09709-1).
Collapse
Affiliation(s)
- Timothy J Vreeland
- Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, MD, USA. .,Department of Surgical Oncology, Brooke Army Medical Center, San Antonio, TX, USA. .,Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Guy T Clifton
- Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, MD, USA.,Department of Surgical Oncology, Brooke Army Medical Center, San Antonio, TX, USA
| | - Diane F Hale
- Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, MD, USA.,Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Robert C Chick
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | | | - Jessica L Cindass
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | - Alexandra M Adams
- Department of Surgery, Brooke Army Medical Center, San Antonio, TX, USA
| | | | | | - Adam C Berger
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
164
|
Xu C, Zhang S, Zhang Y, Tang SQ, Fang XL, Zhu GL, Peng L, Liu JQ, Mao YP, Tang LL, Liu Q, Lin AH, Sun Y, Ma J. Evolving landscape and academic attitudes toward the controversies of global immuno-oncology trials. Int J Cancer 2021; 149:108-118. [PMID: 33544890 PMCID: PMC8248025 DOI: 10.1002/ijc.33503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/19/2022]
Abstract
This cross‐sectional and longitudinal descriptive analysis aimed to track the evolving landscape of global immuno‐oncology (IO) trials and provide insight into the resolution of IO‐related controversies. Clinical trials (n = 4510) registered on ClinicalTrials.gov in 2007 to 2019 studying immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT), cancer vaccines and immune modulators were included. Most of IO trials are Phase 2 and focus on ICIs and multiple IO therapies. The United States leads global IO research, with stable growth and the best methodological quality. Mainland China ranks first in the number of ACT trials but has the lowest article publication rate (6.2%). A multiple‐arm comparative design is often adopted in multiple IO therapies trials (44.0%). Trials studying ICIs and multiple IO therapies are likely to use early registration (80.0% and 86.6%) and stringent corticosteroid‐/infection‐related criteria. Hospitals have provided the most extensive and strongest support for all IO categories. Big pharma prefers to fund Phase 3‐4 ICI trials (6.98%), while small pharma has a wider sponsorship favoring Phase 1‐2 trials. The “partial‐use‐of‐corticosteroids” strategy is generally well accepted in ICI trials with a definitive trend (32.5%; P < .001) but is associated with the poor dissemination of results (P ≤ .020), while the complete disclosure and standardization of dose/timing limits are still lacking. Disparities in design features and dissemination of results are widespread in IO trials and are modulated by IO category, cancer type and sponsor. We propose policy reforms to redefine the timely publication of IO trials and standardize the resolution of corticosteroid‐/infection‐related issues.
What's new?
In recent decades, immunotherapy has emerged and advanced to become a key part of cancer‐fighting strategies. The rapid growth of immuno‐oncology, however, has been accompanied by controversy in suitable interventions and trial design. In this cross‐sectional and longitudinal analysis, disparities in design were found to be common in immuno‐oncology trials, with differences influenced by factors such as cancer type and trial sponsor. Trials with strict limitations on corticosteroid use had significantly higher publications rates than trials permitting partial corticosteroid administration. The data further suggest that timely publication of immuno‐oncology trials is the third year after trial completion.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shu Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.,Sun Yat-sen Global Health Institute, School of Public Health and Institute of State Governance, Sun Yat-sen University, Guangzhou, China
| | - Si-Qi Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xue-Liang Fang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Guang-Li Zhu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liang Peng
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jin-Qi Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qing Liu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ai-Hua Lin
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
165
|
Funck-Brentano E, Malissen N, Roger A, Lebbé C, Deilhes F, Frénard C, Dréno B, Meyer N, Grob JJ, Tétu P, Saiag P. Which adjuvant treatment for patients with BRAF V600-mutant cutaneous melanoma? Ann Dermatol Venereol 2021; 148:145-155. [PMID: 33579557 DOI: 10.1016/j.annder.2020.11.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 12/21/2022]
Abstract
Treatment of patients with melanoma has considerably improved over the past decade and more recently with adjuvant therapies for patients with American Joint Committee on Cancer (AJCC) stage III (loco-regional metastases) or IV (distant metastases) totally resected melanoma, in order to prevent recurrence. In the adjuvant setting, two options are available to patients with BRAFV600-mutant AJCC stage III totally resected melanoma: anti-PD-1 blockers (nivolumab or pembrolizumab) or BRAF plus MEK inhibitors (dabrafenib plus trametinib). In the absence of comparative studies, it is difficult to determine which of these options is best. Our aim was to review published studies focusing on the management of patients with BRAFV600-mutant melanoma in the adjuvant setting. We also reviewed the main clinical trials of BRAF plus MEK inhibitors and immunotherapy in advanced (i.e. unresectable metastatic) BRAF-mutant melanoma in an attempt to identify results potentially affecting the management of patients on adjuvants. More adverse events are observed with targeted therapy, but all resolve rapidly upon drug discontinuation, whereas with immune checkpoint blockers some adverse events may persist. New therapeutic strategies are emerging, notably neoadjuvant therapies for stage III patients and adjuvant therapies for stage II patients; the place of the adjuvant strategy amidst all these options will soon be re-evaluated. The choice of adjuvant treatment could influence the choice of subsequent treatments in neo-adjuvant or metastatic settings. This review will lead clinicians to a better understanding of the different adjuvant treatments available for patients with totally resected AJCC stage III and IV BRAFV600-mutant melanoma before considering subsequent treatment strategies.
Collapse
Affiliation(s)
- E Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, Boulogne-Billancourt, France; Research unit EA4340 "Biomarkers and clinical trials in oncology and onco-hematology", Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, France.
| | - N Malissen
- Department of Dermatology and Skin Cancer, Aix-Marseille University, AP-HM, Hôpital Timone, Marseille, France
| | - A Roger
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, Boulogne-Billancourt, France; Research unit EA4340 "Biomarkers and clinical trials in oncology and onco-hematology", Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, France
| | - C Lebbé
- Inserm U976, Department of Dermatology, Dermatology, Paris University, Hôpital Saint-Louis, AP-HP, Paris, France
| | - F Deilhes
- Dermatology Department, CHU de Toulouse, Toulouse, France
| | - C Frénard
- Department of Dermatology, CRCINA, CIC1413, CHU de Nantes, université de Nantes, Nantes, France
| | - B Dréno
- Department of Dermatology, CRCINA, CIC1413, CHU de Nantes, université de Nantes, Nantes, France
| | - N Meyer
- Dermatology Department, CHU de Toulouse, Toulouse, France
| | - J-J Grob
- Department of Dermatology and Skin Cancer, Aix-Marseille University, AP-HM, Hôpital Timone, Marseille, France
| | - P Tétu
- Department of Dermatology, CRCINA, CIC1413, CHU de Nantes, université de Nantes, Nantes, France
| | - P Saiag
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, Boulogne-Billancourt, France; Research unit EA4340 "Biomarkers and clinical trials in oncology and onco-hematology", Versailles-Saint-Quentin-en-Yvelines University, Paris-Saclay University, France
| |
Collapse
|
166
|
Prodam F, Caputo M, Mele C, Marzullo P, Aimaretti G. Insights into non-classic and emerging causes of hypopituitarism. Nat Rev Endocrinol 2021; 17:114-129. [PMID: 33247226 DOI: 10.1038/s41574-020-00437-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 12/11/2022]
Abstract
Hypopituitarism is defined as one or more partial or complete pituitary hormone deficiencies, which are related to the anterior and/or posterior gland and can have an onset in childhood or adulthood. The most common aetiology is a sellar or suprasellar lesion, often an adenoma, which causes hypopituitarism due to tumour mass effects, or the effects of surgery and/or radiation therapy. However, other clinical conditions, such as traumatic brain injury, and autoimmune and inflammatory diseases, can result in hypopituitarism, and there are also genetic causes of hypopituitarism. Furthermore, the use of immune checkpoint inhibitors to treat cancer is increasing the risk of hypopituitarism, with a pattern of hormone defects that is different from the classic patterns and depends on mechanisms that are specific for each drug. Moreover, autoantibody production against the pituitary and hypothalamus has been demonstrated in studies investigating the development or worsening of some cases of hypopituitarism. Finally, evidence suggests that posterior pituitary damage can affect oxytocin secretion. The aim of this Review is to summarize current knowledge on non-classic and emerging causes of hypopituitarism, so as to help clinicians improve early identification, avoid life-threatening events and improve the clinical care and quality of life of patients at risk of hypopituitarism.
Collapse
Affiliation(s)
- Flavia Prodam
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Marina Caputo
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Chiara Mele
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
| | - Paolo Marzullo
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy
- Division of General Medicine, I.R.C.C.S. Istituto Auxologico Italiano, Ospedale San Giuseppe, Verbania, Italy
| | - Gianluca Aimaretti
- Endocrinology, Department of Translational Medicine, Università del Piemonte Orientale, Novara, Italy.
| |
Collapse
|
167
|
Abstract
The treatment landscape for patients with advanced melanoma has dramatically improved over the past decade, leading to unprecedented survival. Despite the robust activity of single-agent immune-checkpoint blockade with anti-CTLA-4 or anti-PD-1 agents, and the efficacy of targeted therapies capable of interrupting aberrant signaling resulting from BRAF mutations, the benefit from these therapies is not universal. Advanced understanding of immune and molecular processes underlying melanoma tumorigenesis has demonstrated the promise of combined, multidrug regimens. We discuss the currently available evidence that supports using combinatorial approaches in advanced melanoma treatment and provide insights into promising new combination strategies under investigation.
Collapse
Affiliation(s)
- Rodrigo Ramella Munhoz
- Oncology Center, Hospital Sírio Libanês, Rua Dona Adma Jafet, 91, São Paulo 01308-050, Brazil.
| | - Michael Andrew Postow
- Melanoma Service, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
168
|
Karwacka I, Obołończyk Ł, Kaniuka-Jakubowska S, Sworczak K. The Role of Immunotherapy in the Treatment of Adrenocortical Carcinoma. Biomedicines 2021; 9:biomedicines9020098. [PMID: 33498467 PMCID: PMC7909536 DOI: 10.3390/biomedicines9020098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/30/2020] [Accepted: 01/18/2021] [Indexed: 01/20/2023] Open
Abstract
Adrenocortical carcinoma (ACC) is a rare epithelial neoplasm, with a high tendency for local invasion and distant metastases, with limited treatment options. Surgical treatment is the method of choice. For decades, the mainstay of pharmacological treatment has been the adrenolytic drug mitotane, in combination with chemotherapy. Immunotherapy is the latest revolution in cancer therapy, however preliminary data with single immune checkpoint inhibitors showed a modest activity in ACC patients. The anti-neoplastic activity of immune checkpoint inhibitors such as anti-cytotoxic-T-lymphocyte-associated-antigen 4 (anti-CTLA-4), anti-programmed death-1 (anti-PD-1), and anti-PD-ligand-1 (PD-L1) antibodies in different solid tumors has aroused interest to explore the potential therapeutic effect in ACC as well. Multiple ongoing clinical trials are currently evaluating the role of immune checkpoint inhibitors in ACC (pembrolizumab, combination pembrolizumab and relacorilant, nivolumab, combination nivolumab and ipilimumab). The primary and acquired resistance to immunotherapy continue to counter treatment efficacy. Therefore, attempts are made to combine therapy: anti-PD-1 antibody and anti-CTLA-4 antibody, anti-PD-1 antibody and antagonist of the glucocorticoid receptor. The inhibitors of immune checkpoints would benefit patients with antitumor immunity activated by radiotherapy. Immunotherapy is well tolerated by patients; the most frequently observed side effects are mild. The most common adverse effects of immunotherapy are skin and gastrointestinal disorders. The most common endocrinopathy during anti-CTLA treatment is pituitary inflammation and thyroid disorders.
Collapse
|
169
|
Wu J, Das J, Kalra M, Ratto B. Comparative efficacy of dabrafenib + trametinib versus treatment options for metastatic melanoma in first-line settings. J Comp Eff Res 2021; 10:267-280. [PMID: 33448878 DOI: 10.2217/cer-2020-0249] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: The objective was to systematically review the literature and assess the relative efficacy of agents approved in first-line settings via network meta-analysis. Materials & methods: A literature review was conducted via searching different medical databases. The eligibility criteria included Phase II or III randomized controlled trials that had enrolled treatment-naive adult patients with advanced/metastatic melanoma. Results: The network meta-analysis results suggested that dabrafenib + trametinib significantly prolongs the survival outcomes compared with the monotherapies and had comparable efficacy profile compared with encorafenib + binimetinib and cobimetinib + vemurafenib. In comparison with immunotherapies, the results varied for progression-free survival and overall survival. Conclusion: Long-term survival data of dabrafenib + trametinib establishes the combination as one of the preferred treatment options for previously untreated melanoma patients.
Collapse
Affiliation(s)
- Jing Wu
- School of Pharmaceutical Science & Technology, Tianjin University, PR China
| | - Jaydeep Das
- Novartis Healthcare Pvt Ltd, Hyderabad, India
| | - Manik Kalra
- Novartis Healthcare Pvt Ltd, Hyderabad, India
| | - Barbara Ratto
- Novartis Pharmaceuticals Corporation, NJ 07936-1080, US
| |
Collapse
|
170
|
Rahma OE, Reuss JE, Giobbie-Hurder A, Shoja E Razavi G, Abu-Shawer O, Mehra P, Gupta S, Simon R, Khleif SN. Early 3+3 Trial Dose-Escalation Phase I Clinical Trial Design and Suitability for Immune Checkpoint Inhibitors. Clin Cancer Res 2021; 27:485-491. [PMID: 33082209 DOI: 10.1158/1078-0432.ccr-20-2669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/21/2020] [Accepted: 10/16/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Despite the expansion of immune checkpoint inhibitor (ICI) indications, the relationship between ICI dose and toxicity or response is not well established. To understand this correlation, we performed a meta-analysis of ICI trials that used dose escalation. EXPERIMENTAL DESIGN We searched PubMed and abstracts presented at (inter)national meetings for trials using FDA-approved ICIs. The reported rates of grade 3-5 adverse events (G3-5 AE), immune-related adverse events (irAE), and response were correlated with doses within each ICI using marginal exact generalized linear models. RESULTS A total of 74 trials (7,469 patients) published between January 2010 and January 2017 were included. For ipilimumab, the incidence of G3-5 AEs was 34% with a significant 27% reduced risk in lower doses (P = 0.002). However, no relationship was observed between dose and irAEs or response. For nivolumab, the incidence of G3-5 AEs was 20.1% which was lower in non-small cell lung cancer (NSCLC) compared with renal cell carcinoma (RCC) or melanoma (P ≤ 0.05) with no dose-toxicity relationship. In melanoma and NSCLC, a dose-response association was observed, which was not observed in RCC. For pembrolizumab, the incidence of G3-5 AEs was 13.3%, which was lower in melanoma compared with NSCLC (P = 0.03) with no dose-toxicity relationship. In melanoma, lower dose levels correlated with decreased odds of response (P = 0.01), a relationship that was not observed in NSCLC. CONCLUSIONS Our analysis shows a lack of consistent dose-toxicity or dose-response correlation with ICIs. Therefore, dose escalation is not an appropriate design to conduct ICI studies. Here we present an innovative trial design for immune-modulating agents.
Collapse
Affiliation(s)
- Osama E Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Joshua E Reuss
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anita Giobbie-Hurder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | - Pooja Mehra
- University of Virginia, Charlottesville, Virginia
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | | | - Samir N Khleif
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.
| |
Collapse
|
171
|
Hu C, Huang Y, Luo P, Yang Y. Effect of antioxidants coenzyme Q10 and β-carotene on the cytotoxicity of vemurafenib against human malignant melanoma. Oncol Lett 2021; 21:208. [PMID: 33574947 PMCID: PMC7816282 DOI: 10.3892/ol.2021.12469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/13/2020] [Indexed: 11/09/2022] Open
Abstract
Melanoma is a type of highly invasive skin cancer derived from melanocytes with poor prognosis. Vemurafenib (PLX4032) is a clinically approved targeted therapeutic for BRAF mutant melanoma that has a high therapeutic response rate and significantly prolongs the overall survival time of patients with melanoma. Antioxidants have been widely used as supplements for cancer prevention and for decreasing the side effects of cancer therapy. However, antioxidants can also protect cancer cells from oxidative stress and promote cancer growth and progression. The present study aimed to examine the effect of the antioxidants coenzyme Q10 (CoQ10) and β-carotene on melanoma cell growth and invasiveness and on the cytotoxicity of vemurafenib against both vemurafenib-sensitive (SK-MEL-28) and vemurafenib-resistant (A2058) human malignant melanoma cell lines. MTS assay and wound-healing assay demonstrated that CoQ10 alone significantly reduced the viability and migration of melanoma cells, respectively, and synergistically worked with vemurafenib to decrease the viability and migration of human melanoma cells. In contrast, MTS assay and flow cytometry revealed that β-carotene alone did not affect the viability and apoptosis induction of melanoma cells; however, it inhibited cell migration and invasiveness. Wound-healing and Transwell assay demonstrated that β-carotene alleviated the cytotoxicity of vemurafenib and mitigated the inhibitory effect of vemurafenib on cell migration and invasion. Both CoQ10 and β-carotene protected melanoma cells from undergoing apoptosis induced by vemurafenib. Immunoblotting demonstrated that β-carotene at physiological concentration worked synergistically with vemurafenib to suppress the Ras-Raf-Mek-Erk intracellular signaling pathway. The present study aimed to add to the evidence of the in vitro effects of CoQ10 and β-carotene on the antimelanoma effects of vemurafenib.
Collapse
Affiliation(s)
- Changkun Hu
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Yuan Huang
- Department of Hematopathology, Anqing Municipal Hospital, Anqing, Anhui 246004, P.R. China
| | - Peixiao Luo
- School of Natural Sciences, College of Science and Technology, Wenzhou Kean University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yixin Yang
- School of Natural Sciences, College of Science and Technology, Wenzhou Kean University, Wenzhou, Zhejiang 325035, P.R. China.,School of Natural Sciences, The Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, Union, NJ 07083, USA
| |
Collapse
|
172
|
Yang Y, Liu J, Yang K, Ma Y, Fu S, Tang X, Wang Y, Zhou L. Endocrine Adverse Events Caused by Different Types and Different Doses of Immune Checkpoint Inhibitors in the Treatment of Solid Tumors: A Meta-Analysis and Systematic Review. J Clin Pharmacol 2021; 61:282-297. [PMID: 33345342 DOI: 10.1002/jcph.1804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022]
Abstract
The aim of this meta-analysis was to assess the risks of endocrine adverse events in patients with malignancies treated with different types and different doses of immune checkpoint inhibitors (ICIs). PubMed and Embase were searched for randomized controlled trials on ICIs and endocrine adverse events since 2000, and meta-analysis was carried out. Twenty-six randomized controlled trials comprising 13 824 patients with malignancies were included. Compared with the other tumor therapies (used as a control group), patients treated with programmed death-1 inhibitors appeared to be at higher risks of hypothyroidism, hyperthyroidism, thyroiditis, hypophysitis or hypopituitarism, and type 1 diabetes mellitus, while there was no difference in the risk of primary adrenal insufficiency. It was also found that patients treated with cytotoxic T-lymphocyte-associated protein-4 inhibitors were at higher risk of hypophysitis or hypopituitarism, primary adrenal insufficiency, and hypothyroidism. In comparison, patients treated with programmed death-ligand 1 inhibitors were at higher risk of hyperthyroidism and hypothyroidism. Compared with the control group, both low-dose and high-dose ICI groups were at higher risk of hypothyroidism and hyperthyroidism, and the low-dose group had increased risk of thyroiditis and primary adrenal insufficiency. There was no significant difference in the risk of type 1 diabetes between the low-dose group and the high-dose group. The risk of hypophysitis or hypopituitarism in the high-dose group (relative risk, 20.12; 95% confidence interval, 8.02-50.46) was significantly higher than that in the low-dose group (relative risk, 4.92; 95% confidence interval, 2.11-11.47). The risk of endocrine adverse events was increased in patients treated with ICIs. Different types and doses of ICIs have varying characteristics of endocrine adverse events.
Collapse
Affiliation(s)
- Yaxian Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Jingfang Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,Department of Endocrinology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Kaili Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yanqi Ma
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Songbo Fu
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,Department of Endocrinology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xulei Tang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China.,Department of Endocrinology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yan Wang
- Department of Endocrinology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Liyuan Zhou
- Department of Endocrinology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
173
|
Dovedi SJ, Elder MJ, Yang C, Sitnikova SI, Irving L, Hansen A, Hair J, Jones DC, Hasani S, Wang B, Im SA, Tran B, Subramaniam DS, Gainer SD, Vashisht K, Lewis A, Jin X, Kentner S, Mulgrew K, Wang Y, Overstreet MG, Dodgson J, Wu Y, Palazon A, Morrow M, Rainey GJ, Browne GJ, Neal F, Murray TV, Toloczko AD, Dall'Acqua W, Achour I, Freeman DJ, Wilkinson RW, Mazor Y. Design and Efficacy of a Monovalent Bispecific PD-1/CTLA4 Antibody That Enhances CTLA4 Blockade on PD-1 + Activated T Cells. Cancer Discov 2021; 11:1100-1117. [PMID: 33419761 DOI: 10.1158/2159-8290.cd-20-1445] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/04/2020] [Accepted: 12/17/2020] [Indexed: 11/16/2022]
Abstract
The clinical benefit of PD-1 blockade can be improved by combination with CTLA4 inhibition but is commensurate with significant immune-related adverse events suboptimally limiting the doses of anti-CTLA4 mAb that can be used. MEDI5752 is a monovalent bispecific antibody designed to suppress the PD-1 pathway and provide modulated CTLA4 inhibition favoring enhanced blockade on PD-1+ activated T cells. We show that MEDI5752 preferentially saturates CTLA4 on PD-1+ T cells versus PD-1- T cells, reducing the dose required to elicit IL2 secretion. Unlike conventional PD-1/CTLA4 mAbs, MEDI5752 leads to the rapid internalization and degradation of PD-1. Moreover, we show that MEDI5752 preferentially localizes and accumulates in tumors providing enhanced activity when compared with a combination of mAbs targeting PD-1 and CTLA4 in vivo. Following treatment with MEDI5752, robust partial responses were observed in two patients with advanced solid tumors. MEDI5752 represents a novel immunotherapy engineered to preferentially inhibit CTLA4 on PD-1+ T cells. SIGNIFICANCE: The unique characteristics of MEDI5752 represent a novel immunotherapy engineered to direct CTLA4 inhibition to PD-1+ T cells with the potential for differentiated activity when compared with current conventional mAb combination strategies targeting PD-1 and CTLA4. This molecule therefore represents a step forward in the rational design of cancer immunotherapy.See related commentary by Burton and Tawbi, p. 1008.This article is highlighted in the In This Issue feature, p. 995.
Collapse
Affiliation(s)
- Simon J Dovedi
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom.
| | | | - Chunning Yang
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | | | - Lorraine Irving
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Anna Hansen
- Translational Science and Experimental Medicine, Respiratory and Immunology (RI), Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - James Hair
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Des C Jones
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Sumati Hasani
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Bo Wang
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Seock-Ah Im
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, Korea
| | - Ben Tran
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | | | - Kapil Vashisht
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Arthur Lewis
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Xiaofang Jin
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Stacy Kentner
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kathy Mulgrew
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | - Yaya Wang
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | - James Dodgson
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Yanli Wu
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Asis Palazon
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Gareth J Browne
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Frances Neal
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Thomas V Murray
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Aleksandra D Toloczko
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - William Dall'Acqua
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland
| | - Ikbel Achour
- Early Oncology R&D, AstraZeneca, Cambridge, United Kingdom
| | | | | | - Yariv Mazor
- Antibody Discovery and Protein Engineering, R&D, AstraZeneca, Gaithersburg, Maryland.
| |
Collapse
|
174
|
Sangro B, Sarobe P, Hervás-Stubbs S, Melero I. Advances in immunotherapy for hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2021; 18:525-543. [PMID: 33850328 PMCID: PMC8042636 DOI: 10.1038/s41575-021-00438-0] [Citation(s) in RCA: 828] [Impact Index Per Article: 207.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent disease with a progression that is modulated by the immune system. Systemic therapy is used in the advanced stage and until 2017 consisted only of antiangiogenic tyrosine kinase inhibitors (TKIs). Immunotherapy with checkpoint inhibitors has shown strong anti-tumour activity in a subset of patients and the combination of the anti-PDL1 antibody atezolizumab and the VEGF-neutralizing antibody bevacizumab has or will soon become the standard of care as a first-line therapy for HCC, whereas the anti-PD1 agents nivolumab and pembrolizumab are used after TKIs in several regions. Other immune strategies such as adoptive T-cell transfer, vaccination or virotherapy have not yet demonstrated consistent clinical activity. Major unmet challenges in HCC checkpoint immunotherapy are the discovery and validation of predictive biomarkers, advancing treatment to earlier stages of the disease, applying the treatment to patients with liver dysfunction and the discovery of more effective combinatorial or sequential approaches. Combinations with other systemic or local treatments are perceived as the most promising opportunities in HCC and some are already under evaluation in large-scale clinical trials. This Review provides up-to-date information on the best use of currently available immunotherapies in HCC and the therapeutic strategies under development.
Collapse
Affiliation(s)
- Bruno Sangro
- grid.411730.00000 0001 2191 685XLiver Unit and HPB Oncology Area, Clinica Universidad de Navarra-IDISNA and CIBEREHD, Pamplona, Spain
| | - Pablo Sarobe
- grid.5924.a0000000419370271Program of Immunology and Immunotherapy, CIMA de la Universidad de Navarra, IDISNA and CIBEREHD, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- grid.5924.a0000000419370271Program of Immunology and Immunotherapy, CIMA de la Universidad de Navarra, IDISNA and CIBEREHD, Pamplona, Spain
| | - Ignacio Melero
- grid.5924.a0000000419370271Program of Immunology and Immunotherapy, CIMA de la Universidad de Navarra, IDISNA and CIBEREHD, Pamplona, Spain ,grid.411730.00000 0001 2191 685XDepartment of Immunology and Immunotherapy, Clinica Universidad de Navarra-IDISNA and CIBERONC, Pamplona, Spain
| |
Collapse
|
175
|
George AS, Department of Medical Oncology, Lakeshore Hospital, Cochin, Kerala, India, Fernandez CJ, Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK, Eapen D, Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK, Pappachan JM, Department of Endocrinology & Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston, UK, Manchester Metropolitan University, Manchester, UK, The University of Manchester, Manchester, UK. Organ-specific Adverse Events of Immune Checkpoint Inhibitor Therapy, with Special Reference to Endocrinopathies. EUROPEAN ENDOCRINOLOGY 2021; 1:21. [DOI: 10.17925/ee.2021.1.1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
176
|
Zhao Z, Zhang C, Zhou L, Dong P, Shi L. Immune Checkpoint Inhibitors and Neurotoxicity. Curr Neuropharmacol 2021; 19:1246-1263. [PMID: 33380303 PMCID: PMC8719293 DOI: 10.2174/1570159x19666201230151224] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/13/2020] [Accepted: 12/08/2020] [Indexed: 11/22/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have recently been used as a promising treatment for cancer, while their toxicity and immune-related side effects can be seen in any organ, including the nervous system. In contrast to other immune-related adverse events (irAEs), neurological irAEs (nAEs) are rare, with varying incidence and symptom complexity. Although nAEs are uncommon, they can sometimes be severe and even lead to death. However, little attention has been paid to nAEs, and the literature is mostly clinical reports with only a few cases. We, therefore, conducted the present review with the aim of providing a comprehensive introduction of nAEs. In this review, we summarized various nAEs, including meningitis, encephalitis, and hypophysitis in the central nervous system, and myositis, myasthenia gravis, and peripheral neuropathies in the peripheral system. We also reviewed the current diagnosis and treatment methods for nAEs commonly used in clinical practice. In addition, we discussed potential mechanisms regarding nAEs and proposed the possible approaches to prevent the risk of nAEs in patients treated with ICIs. There is still a lot to learn, such as whether and why patients with nAEs respond better to ICI-therapy. The mechanisms and significance of nAEs need to be fully clarified to address these issues and optimize the treatment strategy.
Collapse
Affiliation(s)
| | | | | | - Pan Dong
- Address correspondence to these authors at the School of Life Sciences, Chongqing University, NO.55, University City South Rd, Shapingba District, Chongqing, 400044, China; E-mail: and E-mail:
| | - Lei Shi
- Address correspondence to these authors at the School of Life Sciences, Chongqing University, NO.55, University City South Rd, Shapingba District, Chongqing, 400044, China; E-mail: and E-mail:
| |
Collapse
|
177
|
Sznol M, Melero I. Revisiting anti-CTLA-4 antibodies in combination with PD-1 blockade for cancer immunotherapy. Ann Oncol 2020; 32:295-297. [PMID: 33307201 DOI: 10.1016/j.annonc.2020.11.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 11/24/2022] Open
Affiliation(s)
- M Sznol
- Departments of Immunology and Oncology, Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| | - I Melero
- Department of Immunology, CIMA and Clinica Universidad de Navarra, Pamplona, Spain; CIBERONC, Madrid, Spain
| |
Collapse
|
178
|
Kawakatsu S, Bruno R, Kågedal M, Li C, Girish S, Joshi A, Wu B. Confounding factors in exposure-response analyses and mitigation strategies for monoclonal antibodies in oncology. Br J Clin Pharmacol 2020; 87:2493-2501. [PMID: 33217012 DOI: 10.1111/bcp.14662] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 12/29/2022] Open
Abstract
Dose selection and optimization is an important topic in drug development to maximize treatment benefits for all patients. While exposure-response (E-R) analysis is a useful method to inform dose-selection strategy, in oncology, special considerations for prognostic factors are needed due to their potential to confound the E-R analysis for monoclonal antibodies. The current review focuses on 3 different approaches to mitigate the confounding effects for monoclonal antibodies in oncology: (i) Cox-proportional hazards modelling and case-matching; (ii) tumour growth inhibition-overall survival modelling; and (iii) multiple dose level study design. In the presence of confounding effects, studying multiple dose levels may be required to reveal the true E-R relationship. However, it is impractical for pivotal trials in oncology drug development programmes. Therefore, the strengths and weaknesses of the other 2 approaches are considered, and the favourable utility of tumour growth inhibition-overall survival modelling to address confounding in E-R analyses is described. In the broader scope of oncology drug development, this review discusses the downfall of the current emphasis on E-R analyses using data from single dose level trials and proposes that development programmes be designed to study more dose levels in earlier trials.
Collapse
Affiliation(s)
- Sonoko Kawakatsu
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA.,Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, USA
| | - René Bruno
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA
| | - Matts Kågedal
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA
| | - Chunze Li
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA
| | - Sandhya Girish
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA
| | - Amita Joshi
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA
| | - Benjamin Wu
- Clinical Pharmacology, Development Sciences, gRED, Genentech/Roche, South San Francisco, CA, USA
| |
Collapse
|
179
|
Seth R, Messersmith H, Kaur V, Kirkwood JM, Kudchadkar R, McQuade JL, Provenzano A, Swami U, Weber J, Alluri KC, Agarwala S, Ascierto PA, Atkins MB, Davis N, Ernstoff MS, Faries MB, Gold JS, Guild S, Gyorki DE, Khushalani NI, Meyers MO, Robert C, Santinami M, Sehdev A, Sondak VK, Spurrier G, Tsai KK, van Akkooi A, Funchain P. Systemic Therapy for Melanoma: ASCO Guideline. J Clin Oncol 2020; 38:3947-3970. [PMID: 32228358 DOI: 10.1200/jco.20.00198] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2020] [Indexed: 12/24/2022] Open
Abstract
PURPOSE To provide guidance to clinicians regarding the use of systemic therapy for melanoma. METHODS ASCO convened an Expert Panel and conducted a systematic review of the literature. RESULTS A systematic review, one meta-analysis, and 34 additional randomized trials were identified. The published studies included a wide range of systemic therapies in cutaneous and noncutaneous melanoma. RECOMMENDATIONS In the adjuvant setting, nivolumab or pembrolizumab should be offered to patients with resected stage IIIA/B/C/D BRAF wild-type cutaneous melanoma, while either of those two agents or the combination of dabrafenib and trametinib should be offered in BRAF-mutant disease. No recommendation could be made for or against the use of neoadjuvant therapy in cutaneous melanoma. In the unresectable/metastatic setting, ipilimumab plus nivolumab, nivolumab alone, or pembrolizumab alone should be offered to patients with BRAF wild-type cutaneous melanoma, while those three regimens or combination BRAF/MEK inhibitor therapy with dabrafenib/trametinib, encorafenib/binimetinib, or vemurafenib/cobimetinib should be offered in BRAF-mutant disease. Patients with mucosal melanoma may be offered the same therapies recommended for cutaneous melanoma. No recommendation could be made for or against specific therapy for uveal melanoma. Additional information is available at www.asco.org/melanoma-guidelines.
Collapse
Affiliation(s)
- Rahul Seth
- State University of New York Upstate Medical University, Syracuse, NY
| | | | | | - John M Kirkwood
- University of Pittsburgh School of Medicine, Pittsburgh, PA
- University of Pittsburgh Medical Center, Hillman Cancer Institute, Pittsburgh, PA
| | | | | | | | - Umang Swami
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at New York University, Langone Health, New York, NY
| | | | - Sanjiv Agarwala
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Nancy Davis
- Vanderbilt University Medical Center, Nashville, TN
| | | | - Mark B Faries
- The Angeles Clinic and Research Institute, Los Angeles, CA
- Cedars Sinai Medical Center, Los Angeles, CA
| | - Jason S Gold
- Veterans Administration Boston Healthcare System, West Roxbury, MA
| | | | - David E Gyorki
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Michael O Meyers
- University of North Carolina School of Medicine, Chapel Hill, NC
| | - Caroline Robert
- Gustave Roussy Cancer Centre, Villejuif, France
- Paris-Saclay University, Villejuif, France
| | - Mario Santinami
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Amikar Sehdev
- Indiana University School of Medicine, Indianapolis, IN
| | - Vernon K Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Katy K Tsai
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA
| | | | | |
Collapse
|
180
|
Huang YF, Xie WJ, Fan HY, Du J. Comparative Risks of High-Grade Adverse Events Among FDA-Approved Systemic Therapies in Advanced Melanoma: Systematic Review and Network Meta-Analysis. Front Oncol 2020; 10:571135. [PMID: 33178599 PMCID: PMC7593404 DOI: 10.3389/fonc.2020.571135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/07/2020] [Indexed: 01/03/2023] Open
Abstract
Background: Head-to-head evidence is lacking in comparative risks of high-grade adverse events (AEs) among different systemic treatment options for advanced melanoma. Methods: An up-to-date systematic review and network meta-analysis (NMA) was performed. Randomized controlled trials (RCTs) of patients with advanced melanoma were eligible if at least one intervention was the Food and Drug Administration-approved targeted or immune checkpoint inhibitors. Risks of high-grade AEs were estimated by random-effects Bayesian NMAs, based on relative risks. Surface under the cumulative ranking probabilities was used to assess relative ranking of treatments. The summary incidences were calculated. Results: Twenty-five RCTs (12,925 patients) comparing 10 different systemic treatment options were included. BRAF/MEK had the highest risk of overall high-grade AEs (pooled incidence: 32.11%). BRAF had the highest risk of high-grade arthralgia (0.39%), whereas MEK had the highest risk of high-grade hypertension (2.28%) and nausea (0.37%). Cytotoxic T-lymphocyte antigen 4 (CTLA-4)/chemo had the highest risk of high-grade diarrhea (1.31%), alanine aminotransferase (0.60%), and aspartate aminotransferase elevation (0.59%). Programmed cell death 1 (PD-1)/CTLA-4 had the highest risks of high-grade pyrexia (1.14%) and rash (0.94%). Using PD-1 inhibitor alone had the lowest risks of overall high-grade AEs. Conclusions: Different systemic treatment options have varying high-grade AEs in advanced melanoma treatment. Current evidences highlight the important risks of BRAF/MEK, CTLA-4/chemo, and PD-1/CTLA-4.
Collapse
Affiliation(s)
- Ya-Fang Huang
- School of General Practice and Continuing Education, Capital Medical University, Beijing, China
| | - Wen-Jie Xie
- Department Clinical Research, University of Bern, Bern, Switzerland
| | - Hai-Yu Fan
- Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Juan Du
- School of General Practice and Continuing Education, Capital Medical University, Beijing, China
| |
Collapse
|
181
|
Schwarze JK, Awada G, Cras L, Tijtgat J, Forsyth R, Dufait I, Tuyaerts S, Van Riet I, Neyns B. Intratumoral Combinatorial Administration of CD1c (BDCA-1) + Myeloid Dendritic Cells Plus Ipilimumab and Avelumab in Combination with Intravenous Low-Dose Nivolumab in Patients with Advanced Solid Tumors: A Phase IB Clinical Trial. Vaccines (Basel) 2020; 8:vaccines8040670. [PMID: 33182610 PMCID: PMC7712037 DOI: 10.3390/vaccines8040670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/31/2022] Open
Abstract
Intratumoral (IT) myeloid dendritic cells (myDCs) play a pivotal role in re-licensing antitumor cytotoxic T lymphocytes. IT injection of the IgG1 monoclonal antibodies ipilimumab and avelumab may induce antibody-dependent cellular cytotoxicity, thereby enhancing the release of tumor antigens that can be captured and processed by CD1c (BDCA-1)+ myDCs. Patients with advanced solid tumors after standard care were eligible for IT injections of ≥1 lesion with ipilimumab (10 mg) and avelumab (40 mg) and intravenous (IV) nivolumab (10 mg) on day 1, followed by IT injection of autologous CD1c (BDCA-1)+ myDCs on day 2. IT/IV administration of ipilimumab, avelumab, and nivolumab was repeated bi-weekly. Primary objectives were safety and feasibility. Nine patients were treated with a median of 21 × 106 CD1c (BDCA-1)+ myDCs, and a median of 4 IT/IV administrations of ipilimumab, avelumab, and nivolumab. The treatment was safe with mainly injection-site reactions, but also immune-related pneumonitis (n = 2), colitis (n = 1), and bullous pemphigoid (n = 1). The best response was a durable partial response in a patient with stage IV melanoma who previously progressed on checkpoint inhibitors. Our combinatorial therapeutic approach, including IT injection of CD1c (BDCA-1)+ myDCs, is feasible and safe, and it resulted in encouraging signs of antitumor activity in patients with advanced solid tumors.
Collapse
Affiliation(s)
- Julia Katharina Schwarze
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
- Correspondence: ; Tel.: +32-2-477-64-15
| | - Gil Awada
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Louise Cras
- Department of Anatomopathology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (L.C.); (R.F.)
| | - Jens Tijtgat
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Ramses Forsyth
- Department of Anatomopathology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (L.C.); (R.F.)
| | - Inès Dufait
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Sandra Tuyaerts
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| | - Ivan Van Riet
- Stem Cell Laboratory, Department of Hematology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium;
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (J.T.); (I.D.); (S.T.); (B.N.)
| |
Collapse
|
182
|
Tang SQ, Tang LL, Mao YP, Li WF, Chen L, Zhang Y, Guo Y, Liu Q, Sun Y, Xu C, Ma J. The Pattern of Time to Onset and Resolution of Immune-Related Adverse Events Caused by Immune Checkpoint Inhibitors in Cancer: A Pooled Analysis of 23 Clinical Trials and 8,436 Patients. Cancer Res Treat 2020; 53:339-354. [PMID: 33171025 PMCID: PMC8053869 DOI: 10.4143/crt.2020.790] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The occurrence pattern of immune-related adverse events (irAEs) induced by immune checkpoint inhibitor (ICI) in cancer treatment remains unclear. Materials and Methods Phase II-III clinical trials that evaluated ICI-based treatments in cancer and were published between January 2007 and December 2019 were retrieved from public electronic databases. The pooled median time to onset (PMT-O), resolution (PMT-R), and immune-modulation resolution (PMT-IMR) of irAEs were generated using the metamedian package of R software. RESULTS Twenty-two eligible studies involving 23 clinical trials and 8,436 patients were included. The PMT-O of all-grade irAEs ranged from 2.2 to 14.8 weeks, with the longest in renal events. The PMT-O of grade ≥ 3 irAEs was significantly longer than that of all-grade irAEs induced by programmed cell death protein 1 (PD-1) and its ligand 1 (PD-L1) inhibitors (27.5 weeks vs. 8.4 weeks, p < 0.001) and treatment of nivolumab (NIV) plus ipilimumab (IPI) (7.9 weeks vs. 6.0 weeks, p < 0.001). The PMT-R of all-grade irAEs ranged from 0.1 to 54.3 weeks, with the shortest and longest in hypersensitivity/infusion reaction and endocrine events, respectively. The PMT-IMR of grade ≥ 3 irAEs was significantly shorter than that of all-grade irAEs caused by PD-1/PD-L1 blockade (6.9 weeks vs. 40.6 weeks, p=0.002) and NIV+IPI treatment (3.1 weeks vs. 5.9 weeks, p=0.031). CONCLUSION This study revealed the general and specific occurrence pattern of ICI-induced irAEs in pan-cancers, which was deemed to aid the comprehensive understanding, timely detection, and effective management of ICI-induced irAEs.
Collapse
Affiliation(s)
- Si-Qi Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Wen-Fei Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Lei Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Yuan Zhang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Ying Guo
- Clinical Trials Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Qing Liu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| |
Collapse
|
183
|
Tran KB, Buchanan CM, Shepherd PR. Evolution of Molecular Targets in Melanoma Treatment. Curr Pharm Des 2020; 26:396-414. [PMID: 32000640 DOI: 10.2174/1381612826666200130091318] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 11/21/2019] [Indexed: 12/12/2022]
Abstract
Melanoma is the deadliest type of skin cancers, accounting for more than 80% of skin cancer mortality. Although melanoma was known very early in the history of medicine, treatment for this disease had remained largely the same until very recently. Previous treatment options, including removal surgery and systemic chemotherapy, offered little benefit in extending the survival of melanoma patients. However, the last decade has seen breakthroughs in melanoma treatment, which all emerged following new insight into the oncogenic signaling of melanoma. This paper reviewed the evolution of drug targets for melanoma treatment based on the emergence of novel findings in the molecular signaling of melanoma. One of the findings that are most influential in melanoma treatment is that more than 50% of melanoma tumors contain BRAF mutations. This is fundamental for the development of BRAF inhibitors, which is the first group of drugs that significantly improves the overall survival of melanoma patients compared to the traditional chemotherapeutic dacarbazine. More recently, findings of the role of immune checkpoint molecules such as CTLA-4 and PD1/PD-L1 in melanoma biology have led to the development of a new therapeutic category: immune checkpoint inhibitors, which, for the first time in the history of cancer treatment, produced a durable response in a subset of melanoma patients. However, as this paper discussed next, there is still an unmet need for melanoma treatment. A significant population of patients did not respond to either BRAF inhibitors or immune checkpoint inhibitors. Of those patients who gained an initial response from those therapies, a remarkable percentage would develop drug resistance even when MEK inhibitors were added to the treatment. Finally, this paper discusses some possible targets for melanoma treatment.
Collapse
Affiliation(s)
- Khanh B Tran
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Christina M Buchanan
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.,Auckland Cancer Society Research Centre, University of Auckland, New Zealand
| |
Collapse
|
184
|
Regev A, Avigan MI, Kiazand A, Vierling JM, Lewis JH, Omokaro SO, Di Bisceglie AM, Fontana RJ, Bonkovsky HL, Freston JW, Uetrecht JP, Miller ED, Pehlivanov ND, Haque SA, Harrison MJ, Kullak-Ublick GA, Li H, Patel NN, Patwardhan M, Price KD, Watkins PB, Chalasani NP. Best practices for detection, assessment and management of suspected immune-mediated liver injury caused by immune checkpoint inhibitors during drug development. J Autoimmun 2020; 114:102514. [DOI: 10.1016/j.jaut.2020.102514] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
|
185
|
Barrios DM, Do MH, Phillips GS, Postow MA, Akaike T, Nghiem P, Lacouture ME. Immune checkpoint inhibitors to treat cutaneous malignancies. J Am Acad Dermatol 2020; 83:1239-1253. [PMID: 32461079 PMCID: PMC7572574 DOI: 10.1016/j.jaad.2020.03.131] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/11/2022]
Abstract
As the incidence of cutaneous malignancies continues to rise and their treatment with immunotherapy expands, dermatologists and their patients are more likely to encounter immune checkpoint inhibitors. While the blockade of immune checkpoint target proteins (cytotoxic T-lymphocyte-associated protein-4, programmed cell death-1, and programmed cell death ligand-1) generates an antitumor response in a substantial fraction of patients, there is a critical need for reliable predictive biomarkers and approaches to address refractory disease. The first article of this Continuing Medical Education series reviews the indications, efficacy, safety profile, and evidence supporting checkpoint inhibition as therapeutics for metastatic melanoma, cutaneous squamous cell carcinoma, and Merkel cell carcinoma. Pivotal studies resulting in the approval of ipilimumab, pembrolizumab, nivolumab, cemiplimab, and avelumab by regulatory agencies for various cutaneous malignancies, as well as ongoing clinical research trials, are discussed.
Collapse
Affiliation(s)
- Dulce M Barrios
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mytrang H Do
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medicine, New York, New York
| | - Gregory S Phillips
- State University of New York Downstate Health Sciences University, Brooklyn, New York
| | - Michael A Postow
- Weill Cornell Medicine, New York, New York; Melanoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Tomoko Akaike
- Division of Dermatology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Paul Nghiem
- Division of Dermatology, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Mario E Lacouture
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medicine, New York, New York.
| |
Collapse
|
186
|
Ma Y, Fang W, Zhao H, Bathena SP, Tendolkar A, Sheng J, Zhang L. A Phase I Dose Escalation Study of the Safety, Tolerability, and Pharmacokinetics of Ipilimumab in Chinese Patients with Select Advanced Solid Tumors. Oncologist 2020; 26:e549-e566. [PMID: 33105036 DOI: 10.1002/onco.13577] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/16/2020] [Indexed: 11/06/2022] Open
Abstract
LESSONS LEARNED The overall safety profiles of ipilimumab 3 mg/kg and 10 mg/kg administered every 3 weeks, were consistent between Chinese patients with solid tumors in the current study and patients from previous U.S. ipilimumab monotherapy studies. No new safety signals were identified. The mean systemic exposures to ipilimumab (assessed by first dose area under the curve during the dosing interval and maximum serum concentration) were numerically lower in the Chinese patient population than in U.S. patients for both 3 mg/kg and 10 mg/kg doses; however, the range of serum concentrations in the Chinese and U.S. populations overlapped (3 mg/kg and 10 mg/kg), suggesting that ipilimumab pharmacokinetics was ethnically insensitive in this study. BACKGROUND This phase I, open-label study assessed ipilimumab safety, tolerability, pharmacokinetics (PK), immunogenicity, and antitumor activity in Chinese patients with unresectable, metastatic, recurrent malignant melanoma (MM) or nasopharyngeal carcinoma (NPC). METHODS Of 39 patients enrolled, 25 received ipilimumab (11 patients received 3 mg/kg, and 14 patients received 10 mg/kg). Reasons for not receiving treatment were withdrawal of consent (3 patients), no longer meeting the criteria (10 patients), and one recorded as "other." During the induction phase, patients received ipilimumab (3 mg/kg, i.v.), on day 1 of a 3-week cycle, to a maximum of four doses or progressive disease (PD). During the maintenance phase at week 24, patients received ipilimumab (3 mg/kg, i.v.) on day 1 of a 12-week cycle, to a maximum of 3 years or PD. Considering the co-primary safety and PK endpoints, the successive dosing required nine patients with two or fewer dose-limiting toxicities during the 42-day observation period to proceed with a new cohort of nine patients at 10 mg/kg. RESULTS Ipilimumab safety and PK profiles were similar in Chinese and predominantly White populations. Ipilimumab was well tolerated. Most adverse events (AEs) were grades 1-2 and experienced by 11 patients treated with 3 mg/kg and 14 patients treated with 10 mg/kg. There were no new safety concerns. Incidence of anti-ipilimumab antibodies was low (1 of 10 in the 3 mg/kg patients and 2 of 13 in the 10 mg/kg patients) and without safety implications. In the 3 mg/kg group, 8 of 11 patients had PD. In the 10 mg/kg group (all NPC, 0 MM patients), 11 of 14 patients had PD. Three patients had stable disease (one at 3 mg/kg and two at 10 mg/kg). CONCLUSION Ipilimumab was well tolerated in Chinese patients, showing similar safety and PK to previous studies in predominantly White populations.
Collapse
Affiliation(s)
- Yuxiang Ma
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Sai Praneeth Bathena
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Lawrenceville, New Jersey, USA
| | - Amol Tendolkar
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Lawrenceville, New Jersey, USA
| | - Jennifer Sheng
- Clinical Pharmacology and Pharmacometrics, Bristol-Myers Squibb, Lawrenceville, New Jersey, USA
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, People's Republic of China
| |
Collapse
|
187
|
Cheung VTF, Brain O. Immunotherapy induced enterocolitis and gastritis - What to do and when? Best Pract Res Clin Gastroenterol 2020; 48-49:101703. [PMID: 33317787 DOI: 10.1016/j.bpg.2020.101703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 01/31/2023]
Abstract
Oncological treatment has been revolutionised by the advent of immune checkpoint inhibitors (ICPi), which block inhibitory immune pathways to enhance anti-tumour responses and improve survival. This mode of action is non-specific so can cause immune-related adverse events, of which diarrhoea and enterocolitis are amongst the most common. ICPi-enterocolitis frequently leads to cancer therapy interruption. ICPi-gastritis typically occurs at a later stage of ICPi therapy and can present more insidiously with nausea and vomiting. ICPi-enterocolitis and gastritis are treated with corticosteroids, with refractory cases typically requiring biologic therapy. This review will briefly consider the pathogenesis of ICPi-induced GI disease, before focussing on the practical management of these conditions. The anticipated global increase in ICPi use across cancer types highlights the importance of prospective research in order that we can understand the immuno-microbiology of ICPi-enterocolitis and gastritis. This will lead to predictive biomarkers and help to define optimal treatment regimens.
Collapse
Affiliation(s)
- Vincent Ting Fung Cheung
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | - Oliver Brain
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
188
|
Foo T, Tapia Rico G, Roberts-Thomson R. Immunotherapy in Older Patients with Advanced Melanoma: A Review of the Current Evidence. Drugs Aging 2020; 37:411-423. [PMID: 32307654 DOI: 10.1007/s40266-020-00762-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite the increasing incidence of metastatic melanoma in the older population, there is relatively limited specific data surrounding the use of immunotherapy for the treatment of advanced melanoma for patients above the age of 65 years. To date, there has not been a prospective trial done to evaluate the safety and efficacy of using immunotherapy to treat older patients with advanced melanoma. Older patients are often under-represented in clinical trials. In addition, older patients in clinical trials may have lower Eastern Cooperative Oncology Group (ECOG) performance score and fewer co-morbidities, and thus trial data may not truly reflect the experience of treating older patients. The purpose of this descriptive review is to examine the efficacy and safety data of the three currently approved immune checkpoint inhibitors for advanced melanoma treatment in older patients. Our review of available data established that the efficacy and tolerability of immunotherapy in older patients are comparable to results seen in younger patients. However, a dedicated, prospective, randomised trial to assess the safety, tolerability, and quality-of-life parameters of immunotherapy in the older population would provide further insight on the value of these treatments.
Collapse
Affiliation(s)
- Tiffany Foo
- Adelaide Oncology and Haematology, Adelaide, SA, Australia
| | - Gonzalo Tapia Rico
- Medical Oncology Department, Royal Adelaide Hospital, Adelaide, SA, Australia.
| | - Rachel Roberts-Thomson
- Adelaide Oncology and Haematology, Adelaide, SA, Australia
- Medical Oncology Department, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| |
Collapse
|
189
|
Durrechou Q, Domblides C, Sionneau B, Lefort F, Quivy A, Ravaud A, Gross-Goupil M, Daste A. Management of Immune Checkpoint Inhibitor Toxicities. Cancer Manag Res 2020; 12:9139-9158. [PMID: 33061607 PMCID: PMC7533913 DOI: 10.2147/cmar.s218756] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/07/2020] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have radically changed the clinical outcome of several cancers with durable responses. CTLA-4 (cytotoxic T lymphocyte antigen-4), PD-1 (programmed cell death protein 1) or PDL-1 (programmed cell death ligand protein 1) represent ICIs that can be used as monotherapy or in combination with other agents. The toxicity p\rofiles of ICIs differ from the side effects of cytotoxic agents and come with new toxicities like immune-related adverse events. Typically, these toxicities occur in all organs. However, the main organs affected are the skin, digestive, hepatic, lungs, rheumatologic, and endocrine. Most of the immune toxicity that occurs is low grade but some more severe toxicities can occur that require a rapid diagnosis and appropriate treatment. The recognition of symptoms by physicians and patient is necessary to resolve them rapidly and adapt treatment to allow the toxicity to resolve.
Collapse
Affiliation(s)
- Quentin Durrechou
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| | - Charlotte Domblides
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France.,ImmunoConcEpt, CNRS UMR 5164, Bordeaux University, Bordeaux 33076, France
| | - Baptiste Sionneau
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| | - Felix Lefort
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| | - Amandine Quivy
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| | - Alain Ravaud
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| | - Marine Gross-Goupil
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| | - Amaury Daste
- Department of Medical Oncology, Hôpital Saint-André, CHU Bordeaux-University of Bordeaux, Bordeaux, France
| |
Collapse
|
190
|
Wilson BE, Hansen AR. Dual Checkpoint Blockade in Metastatic Castration-Resistant Prostate Cancer: Just a Gambit or Real CheckMate? Cancer Cell 2020; 38:438-440. [PMID: 33049204 DOI: 10.1016/j.ccell.2020.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this issue of Cancer Cell, Sharma et al. report that ipilimumab and nivolumab combination achieved impressive response rates in patients with metastatic castration-resistant prostate cancer (mCRPC). But this regimen produced high rates of toxicity, treatment-related discontinuation, and death. Tolerability needs improvement for this combination to benefit more men with mCRPC.
Collapse
Affiliation(s)
- Brooke E Wilson
- Princess Margaret Cancer Centre, Division of Medical Oncology, University Health Network, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada; University of New South Wales, Kensington, NSW, Australia
| | - Aaron R Hansen
- Princess Margaret Cancer Centre, Division of Medical Oncology, University Health Network, Toronto, ON, Canada; University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
191
|
Muir CA, Menzies AM, Clifton-Bligh R, Tsang VHM. Thyroid Toxicity Following Immune Checkpoint Inhibitor Treatment in Advanced Cancer. Thyroid 2020; 30:1458-1469. [PMID: 32264785 DOI: 10.1089/thy.2020.0032] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background: Inhibitory antibodies against cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1) have antitumor efficacy and are now standard of care in the management of multiple cancer subtypes. However, the use is complicated by the development of autoimmunity, which can occur in multiple organ systems. Thyroiditis is the most common immune-related adverse event. Summary: Immune checkpoint inhibitor (ICI)-associated thyroiditis affects over 10% of treated patients. PD-1 inhibitors are associated with greater risk of thyroid dysfunction relative to CTLA-4 inhibitors, although the highest risk occurs with combined anti-CTLA-4 and anti-PD-1 treatment. Onset is typically rapid, within weeks to months and both hyperthyroidism and hypothyroidism can occur. The most frequent pattern of thyroid dysfunction is transient hyperthyroidism with evolution to hypothyroidism over four to six weeks. Most cases are asymptomatic and resolve without dedicated treatment. There is no sex or age predominance, and predictive risk factors have not been reliably identified. Thyroid autoantibodies are variably present and are not clearly related to the risk or progression of thyroid dysfunction following treatment with an ICI. Observational data suggest that development of ICI-associated thyroiditis may predict improved survival. Conclusions: ICI-associated thyroiditis is a distinct clinical entity. Mechanisms underlying etiology remain largely unknown. Awareness among health professionals is important to limit morbidity and avoid unnecessary periods of untreated hypothyroidism.
Collapse
Affiliation(s)
- Christopher A Muir
- Nothern Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St. Leonards, Australia
| | - Alexander M Menzies
- Nothern Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Melanoma Medical Oncology, Melanoma Institute Australia, Wollstonecraft, Australia
- Department of Medical Oncology and Royal North Shore Hospital, St. Leonards, Australia
| | - Roderick Clifton-Bligh
- Nothern Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St. Leonards, Australia
- Department of Endocrinology, Royal North Shore Hospital, St. Leonards, Australia
| | - Venessa H M Tsang
- Nothern Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
- Cancer Genetics Laboratory, Kolling Institute of Medical Research, St. Leonards, Australia
- Department of Endocrinology, Royal North Shore Hospital, St. Leonards, Australia
| |
Collapse
|
192
|
Sha CM, Lehrer EJ, Hwang C, Trifiletti DM, Mackley HB, Drabick JJ, Zaorsky NG. Toxicity in combination immune checkpoint inhibitor and radiation therapy: A systematic review and meta-analysis. Radiother Oncol 2020; 151:141-148. [PMID: 32717359 DOI: 10.1016/j.radonc.2020.07.035] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Immune checkpoint inhibitor with radiation therapy (ICI + RT) is under investigation for improved patient outcome, so we performed a systematic review/meta-analysis of toxicities for ICI + RT compared to immune checkpoint inhibitor (ICI) therapy alone. MATERIALS AND METHODS A PRISMA-compliant systematic review of studies in MEDLINE (PubMed) and in the National Comprehensive Cancer Network guidelines was conducted, with primary outcome grade 3 + toxicity. Criteria for ICI alone were: phase III/IV trials that compared immunotherapy to placebo, chemotherapy, or alternative immunotherapy; and for ICI + RT: prospective/retrospective studies with an arm treated with ICI + RT. Meta-analysis was performed by random effects models using the DerSimonian and Laird method. The I2 statistic and Cochran's Q test were used to assess heterogeneity, while funnel plots and Egger's test assessed publication bias. RESULTS This meta-analysis included 51 studies (n = 15,398), with 35 ICI alone (n = 13,956) and 16 ICI + RT studies (n = 1,442). Our models showed comparable grade 3-4 toxicities in ICI + RT (16.3%; 95% CI, 11.1-22.3%) and ICI alone (22.3%; 95% CI, 18.1-26.9%). Stratification by timing of radiation and irradiated site showed no significant differences, but anti-CTLA-4 therapy and melanoma showed increased toxicity. The grade 5 toxicities were 1.1% and 1.9% for ICI alone and ICI + RT respectively. There was significant heterogeneity, but not publication bias. CONCLUSIONS The random effects model showed comparable grade 3-4 toxicity in using ICI + RT compared to ICI alone in CNS melanoma metastases, NSCLC, and prostate cancer. ICI + RT is safe for future clinical trials in these cancers.
Collapse
Affiliation(s)
- Congzhou M Sha
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA
| | - Eric J Lehrer
- Department of Radiation Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clara Hwang
- Department of Internal Medicine, Division of Hematology/Oncology, Henry Ford Health System, Detroit, MI, USA
| | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic Jacksonville, Jacksonville, FL, USA
| | - Heath B Mackley
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA
| | - Joseph J Drabick
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA
| | - Nicholas G Zaorsky
- Department of Radiation Oncology, Penn State Cancer Institute, Hershey, PA, USA; Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
193
|
Bomze D, Asher N, Hasan Ali O, Flatz L, Azoulay D, Markel G, Meirson T. Survival-Inferred Fragility Index of Phase 3 Clinical Trials Evaluating Immune Checkpoint Inhibitors. JAMA Netw Open 2020; 3:e2017675. [PMID: 33095247 PMCID: PMC7584930 DOI: 10.1001/jamanetworkopen.2020.17675] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
IMPORTANCE In science and medical research, extreme and dichotomous conclusions may be drawn based on whether the P value falls above or below the threshold. The fragility index (ie, the minimum number of changes from nonevents to events resulting in loss of statistical significance) captures the vulnerability of statistics in trials with binary outcomes. There are a growing number of clinical trials of immune checkpoint inhibitors (ICIs), as well as expanding eligibility for patients to receive them. The robustness of survival outcomes in randomized clinical trials (RCTs) should be evaluated using the fragility index extended to time-to-event data. OBJECTIVE To calculate the fragility of survival data in RCTs evaluating ICIs. DESIGN, SETTING, AND PARTICIPANTS In this cross-sectional study, data on phase 3 prospective RCTs investigating ICIs included in PubMed from inception until January 1, 2020, were extracted. Two- or three-group studies reporting results for overall survival were eligible for the survival-inferred fragility index (SIFI) calculation, which is the minimum number of reassignments of the best survivors from the interventional group to the control group resulting in loss of significance (defined as P < .05 by log-rank test). For nonsignificant results, a negative SIFI was calculated by reversing the direction of reassignment (from the control group to the interventional group). MAIN OUTCOMES AND MEASURES Survival-inferred fragility index. RESULTS A total of 45 phase 3 prospective RCTs (4 of which had 3 groups, for a total of 49 groups) were identified, of which 6 (13%) investigated anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) agents, 25 (56%) investigated anti-programmed cell death 1 (PD-1) agents, 12 (27%) investigated anti-programmed cell death 1 ligand 1 agents, and 3 (7%) investigated the combination of anti-CTLA-4 and anti-PD-1 agents. The median SIFI was 5 (interquartile range, -4 to 12) for the intention-to-treat analysis; for these trials, the SIFI was 1% or less of the total sample size in 17 of 49 populations (35%). In 25 of the 49 intention-to-treat populations (51%), the SIFI was less than the number of censored patients in the intervention group shortly after randomization (defined as <5% of the follow-up time). CONCLUSIONS AND RELEVANCE This study suggests that many phase 3 RCTs evaluating ICI therapies have a low SIFI for overall survival, resulting in uncertainty regarding their potential clinical benefit. Although not a definitive solution for the problems arising from dichotomization, SIFI provides an additional means of assessing and communicating the strength of statistical conclusions.
Collapse
Affiliation(s)
- David Bomze
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Nethanel Asher
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
| | - Omar Hasan Ali
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Lukas Flatz
- Institute for Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
- Department of Oncology, Kantonsspital St Gallen, St Gallen, Switzerland
| | - Daniel Azoulay
- Center for Liver Diseases, Sheba Medical Center, Ramat-Gan, Israel
| | - Gal Markel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
- Department of Clinical Microbiology and Immunology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tomer Meirson
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat-Gan, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
194
|
Stelmachowska-Banaś M, Czajka-Oraniec I. Management of endocrine immune-related adverse events of immune checkpoint inhibitors: an updated review. Endocr Connect 2020; 9:R207-R228. [PMID: 33064663 PMCID: PMC7576644 DOI: 10.1530/ec-20-0342] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
Immune checkpoint inhibitors (ICIs) belong to a new group of anticancer drugs targeting T-cell proteins involved in the activation of immune response toward malignancies. Their introduction into clinical practice was a milestone in modern cancer treatment. However, the significant advantage of ICIs over conventional chemotherapy in terms of therapeutic efficacy is accompanied by new challenges related to specific side effects. ICI-induced immune system activation could lead to the loss of self-tolerance, presenting as autoimmune inflammation and dysfunction of various tissues and organs. Thus, the typical side effects of ICIs include immune-related adverse events (irAEs), among which endocrine irAEs, affecting numerous endocrine glands, have been commonly recognized. This review aimed to outline the current knowledge regarding ICI-induced endocrine disorders from a clinical perspective. We present updated information on the incidence and clinical development of ICI-induced endocrinopathies, including the most frequent thyroiditis and hypophysitis, the rarely observed insulin-dependent diabetes mellitus and primary adrenal insufficiency, and the recently described cases of hypoparathyroidism and lipodystrophy. Practical guidelines for monitoring, diagnosis, and treatment of ICI-related endocrine toxicities are also offered. Rising awareness of endocrine irAEs among oncologists, endocrinologists, and other health professionals caring for patients receiving ICIs could contribute to better safety and efficacy. As immunotherapy becomes widespread and approved for new types of malignancies, increased incidences of endocrine irAEs are expected in the future.
Collapse
Affiliation(s)
- Maria Stelmachowska-Banaś
- Department of Endocrinology, The Centre of Postgraduate Medical Education, Warsaw, Polska, Poland
- Correspondence should be addressed to M Stelmachowska-Banaś:
| | - Izabella Czajka-Oraniec
- Department of Endocrinology, The Centre of Postgraduate Medical Education, Warsaw, Polska, Poland
| |
Collapse
|
195
|
Durbin SM, Mooradian MJ, Fintelmann FJ, Zubiri L, Chute DF, Kambadakone A, Pisuchpen N, Reynolds KL, Dougan M. Diagnostic utility of CT for suspected immune checkpoint inhibitor enterocolitis. J Immunother Cancer 2020; 8:e001329. [PMID: 33033184 PMCID: PMC7545618 DOI: 10.1136/jitc-2020-001329] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND AIMS Immune checkpoint inhibitor (ICI) enterocolitis is a common immune-related adverse event and can be fatal, especially when not diagnosed and treated promptly. The current gold standard for diagnosis is endoscopy with biopsy, but CT scan is a possible alternative. The primary objective of this study is to identify the diagnostic performance of CT in the evaluation of ICI enterocolitis. METHODS With institutional review board approval, we conducted a retrospective cohort study of patients who received ICI therapy between 2015 and 2019 across a healthcare system. Patients were included if they underwent both abdominal CT and endoscopy with biopsy within 3 days. The radiological and pathological diagnoses, as well as clinical characteristics, were extracted from the electronic medical record. We calculated the sensitivity, specificity, positive predictive value (PPV) and negative predictive value (NPV) of CT for diagnosing ICI enterocolitis when compared with tissue diagnosis. RESULTS Of the 4474 patients screened, 138 met inclusion criteria. Most common tumor types were melanoma (37%) and lung cancer (19%). Seventy-four per cent were treated with antiprogrammed cell death (PD-1)/PD-L1 therapy. Thirty-nine per cent had signs of enterocolitis on CT scan and 58% had biopsy-proven ICI enterocolitis. Sensitivity and specificity of CT were 50% and 74%, respectively. PPV was 73% and NPV was 52%. Of those with confirmed ICI enterocolitis, 70% had grade 3 or higher symptoms, 91% received steroids and 40% received infliximab. CONCLUSION The performance of CT scan for diagnosis of ICI enterocolitis is moderate to poor and does not replace endoscopy with biopsy.
Collapse
Affiliation(s)
- Sienna M Durbin
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Meghan J Mooradian
- Harvard Medical School, Boston, Massachusetts, USA
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Florian Johannes Fintelmann
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Leyre Zubiri
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Donald F Chute
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Avinash Kambadakone
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nisanard Pisuchpen
- Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- The Thai Red Cross Society, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kerry L Reynolds
- Harvard Medical School, Boston, Massachusetts, USA
- Cancer Center, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael Dougan
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
196
|
Michielin O, Atkins MB, Koon HB, Dummer R, Ascierto PA. Evolving impact of long-term survival results on metastatic melanoma treatment. J Immunother Cancer 2020; 8:e000948. [PMID: 33037115 PMCID: PMC7549477 DOI: 10.1136/jitc-2020-000948] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Melanoma treatment has been revolutionized over the past decade. Long-term results with immuno-oncology (I-O) agents and targeted therapies are providing evidence of durable survival for a substantial number of patients. These results have prompted consideration of how best to define long-term benefit and cure. Now more than ever, oncologists should be aware of the long-term outcomes demonstrated with these newer agents and their relevance to treatment decision-making. As the first tumor type for which I-O agents were approved, melanoma has served as a model for other diseases. Accordingly, discussions regarding the value and impact of long-term survival data in patients with melanoma may be relevant in the future to other tumor types. Current findings indicate that, depending on the treatment, over 50% of patients with melanoma may gain durable survival benefit. The best survival outcomes are generally observed in patients with favorable prognostic factors, particularly normal baseline lactate dehydrogenase and/or a low volume of disease. Survival curves from melanoma clinical studies show a plateau at 3 to 4 years, suggesting that patients who are alive at the 3-year landmark (especially in cases in which treatment had been stopped) will likely experience prolonged cancer remission. Quality-of-life and mixture-cure modeling data, as well as metrics such as treatment-free survival, are helping to define the value of this long-term survival. In this review, we describe the current treatment landscape for melanoma and discuss the long-term survival data with immunotherapies and targeted therapies, discussing how to best evaluate the value of long-term survival. We propose that some patients might be considered functionally cured if they have responded to treatment and remained treatment-free for at least 2 years without disease progression. Finally, we consider that, while there have been major advances in the treatment of melanoma in the past decade, there remains a need to improve outcomes for the patients with melanoma who do not experience durable survival.
Collapse
Affiliation(s)
- Olivier Michielin
- Oncology Department, Precision Oncology Center, Lausanne, Switzerland
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Michael B Atkins
- Medical Oncology, Georgetown Lombardi Comprehensive Cancer Center and Oncology Academic Department, Georgetown University Medical Center, Washington, DC, USA
| | - Henry B Koon
- Clinical Research, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Paolo Antonio Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Naples, Italy
| |
Collapse
|
197
|
Champiat S, Tselikas L, Farhane S, Raoult T, Texier M, Lanoy E, Massard C, Robert C, Ammari S, De Baère T, Marabelle A. Intratumoral Immunotherapy: From Trial Design to Clinical Practice. Clin Cancer Res 2020; 27:665-679. [PMID: 32943460 DOI: 10.1158/1078-0432.ccr-20-0473] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/10/2020] [Accepted: 09/14/2020] [Indexed: 11/16/2022]
Abstract
Systemic immunotherapies such as immune checkpoint blockade targeted at PD(L)1 and CTLA4 have demonstrated their ability to provide durable tumor responses and long-term overall survival benefits for some patients in several solid tumor types. However, a majority of patients remain resistant to these treatments and a significant proportion of them develop severe autoimmune and inflammatory adverse events. Preclinical studies have demonstrated that intratumoral injections of immunostimulatory products (oncolytics, pattern recognition receptor agonists,…) that are able to trigger type I IFN release and enhance tumor antigen presentation on immune cells could generate a strong antitumor immunity and overcome the resistance to systemic immune checkpoint blockade therapies. The intratumoral immunotherapy strategies that are currently in clinical development offer a unique therapeutic and exploratory setting to better understand the immune contexture across tumor lesions of patients with metastatic cancer. Also these local therapeutic products could turn cold tumors into hot and improve the response rates to cancer immunotherapies while diminishing their systemic exposure and toxicities. Intratumoral immunotherapies could prime or boost the immunity against tumors and therefore radically change the combinatorial therapeutic strategies currently pursued for metastatic and local cancers to improve their long-term survival. We aimed to review and discuss the scientific rationale for intratumoral immunotherapy, the challenges raised by this strategy in terms of drug development within clinical trials and the current state-of-the-art regarding the clinical practice of this innovative approach.
Collapse
Affiliation(s)
- Stéphane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France
| | - Lambros Tselikas
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Siham Farhane
- Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thibault Raoult
- Service de Promotion des Etudes Cliniques (SPEC), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Matthieu Texier
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Emilie Lanoy
- Service de Biostatistiques et d'Epidémiologie (SBE), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Christophe Massard
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Robert
- Département de Médecine Oncologique (DMO), Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Samy Ammari
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Thierry De Baère
- Département de Radiologie, Gustave Roussy, Université Paris Saclay, Villejuif, France.,Université Paris Saclay, Saint-Aubin, France
| | - Aurélien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France. .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), Gustave Roussy, Villejuif, France.,Gustave Roussy Immunotherapy Program (GRIP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| |
Collapse
|
198
|
Improved Progression-Free Long-Term Survival of a Nation-Wide Patient Population with Metastatic Melanoma. Cancers (Basel) 2020; 12:cancers12092591. [PMID: 32932758 PMCID: PMC7564087 DOI: 10.3390/cancers12092591] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/02/2022] Open
Abstract
Simple Summary New cancer treatments have prolonged the lives for patients with metastatic melanoma in clinical trials. However, patients in clinical trials often presents with a better prognosis than other patients. It is therefore important to examine the effect of the new treatments in a real-life setting. We found that survival for patients with metastatic melanoma in a nation-wide setting were prolonged and a higher proportion of these patients were alive and without active disease 3 years after they started treatment. Abstract Approval of immune checkpoint-inhibitors (ICIs) and BRAF-inhibitors has revolutionized the treatment of metastatic melanoma. Although these drugs have improved overall survival (OS) in clinical trials, real-world evidence for improved long-term survival is still scarce. Clinical data were extracted from the Danish Metastatic Melanoma database. This nation-wide cohort contains data on all patients who received systemic treatment for metastatic melanoma between 2008 and 2016. Ipilimumab, the first approved ICI, was implemented as standard-of-care in Denmark in 2012. Hence, patients were divided in a pre-ICI (2008–2011) and an ICI (2012–2016) era. Patients were defined as long-term survivors if they were alive 3 years after initiation of systemic therapy. Data from 1754 patients were retrieved. Patients treated in the ICI era had an improved median OS (11.3 months, 95% confidence interval (CI) 10.3–12.3) compared with those in the pre-ICI era (median OS 8.3 months, 95% CI 7.4–9.5, p < 0.0001). A higher proportion of long-term survivors was observed in the ICI era (survivors >3 years increased from 13% to 26% and survivors >5 years increased from 9% to 21%; both p < 0.0001). For long-term survivors, known prognostic factors were equally distributed between the two periods, except that long-term survivors in the pre-ICI era were younger. For long-term survivors, 70% were without progression in the ICI era compared with 43% in the pre-ICI era (p < 0.0001). For all patients, the proportion without progression increased from 5% to 18% between the pre-ICI and the ICI era (p < 0.0001), respectively. Implementation of ICI has led to a significant increase in progression-free, long-term survival for real-life patients with metastatic melanoma.
Collapse
|
199
|
Abstract
Malignant melanoma is an aggressive skin cancer that originates from cells of the melanocytic lineage and is associated with an invasive growth pattern and early spread. Besides endogenous risk factors such as fair skin type or genetic disposition for the formation of multiple nevi, exposure to ultraviolet light is the most important exogenous risk factor. Treatment of patients with primary tumors includes the complete excision of the primary lesion with appropriate safety margins and in patients with an increased risk of metastasis sentinel lymph node excision. Prognostically significant parameters are the Breslow invasion depth, ulceration of the primary lesion, and sentinel lymph node status. Systemic therapy plays an important role in the adjuvant setting and for inoperable tumors. Depending on the indication and the molecular profile of the tumor tissue, immune checkpoint inhibitors or targeted kinase inhibitors can be used and may result in a significant prolongation of survival times.
Collapse
|
200
|
Chua CYX, Ho J, Demaria S, Ferrari M, Grattoni A. Emerging technologies for local cancer treatment. ADVANCED THERAPEUTICS 2020; 3:2000027. [PMID: 33072860 PMCID: PMC7567411 DOI: 10.1002/adtp.202000027] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Indexed: 12/13/2022]
Abstract
The fundamental limitations of systemic therapeutic administration have prompted the development of local drug delivery platforms as a solution to increase effectiveness and reduce side effects. By confining therapeutics to the site of disease, local delivery technologies can enhance therapeutic index. This review highlights recent advances and opportunities in local drug delivery strategies for cancer treatment in addition to challenges that need to be addressed to facilitate clinical translation. The benefits of local cancer treatment combined with technological advancements and increased understanding of the tumor microenvironment, present a prime breakthrough opportunity for safer and more effective therapies.
Collapse
Affiliation(s)
- Corrine Ying Xuan Chua
- Department of Nanomedicine, Houston Methodist Research Institute (HMRI), Houston, TX, 77030, USA
| | - Jeremy Ho
- Department of Nanomedicine, Houston Methodist Research Institute (HMRI), Houston, TX, 77030, USA
- School of Medicine, Weill Cornell Medical College, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mauro Ferrari
- University of Washington, Box 357630, H375 Health Science Building, Seattle, WA, 98195, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute (HMRI), Houston, TX, 77030, USA
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, 77030, USA
- Department of Surgery, Houston Methodist Hospital, Houston, TX, 77030, USA
| |
Collapse
|