101
|
Krattinger R, Ramelyte E, Dornbierer J, Dummer R. Is single versus combination therapy problematic in the treatment of cutaneous melanoma? Expert Rev Clin Pharmacol 2020; 14:9-23. [PMID: 31364890 DOI: 10.1080/17512433.2019.1650641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: The development of immunotherapies and targeted therapies has changed the treatment approach in resectable, nonresectable, and metastatic melanoma. Because of their different pharmacological profiles, immunotherapies and/or targeted therapies have been studied in various combinations. Areas covered: We reviewed PubMed for most important clinical trials investigating efficacy and tolerability of combinatorial and single-agent approaches for the treatment of melanoma that were published up to June 2019. We discuss the most promising therapy approaches and highlight challenges of melanoma treatment. Expert opinion: Combinatorial approaches seem to be very promising in the treatment of resectable and advanced melanoma. Currently, dual immune checkpoint inhibition (ICI) with nivolumab and ipilimumab offers the best first-line treatment option for patients with BRAF-wt and -mutated, advanced melanoma. It is therapy of choice in younger patients with good ECOG performance status and poor prognostic features, whereas ICI monotherapy should be preferred in elderly patients with advanced melanoma. Benefit-risk ratio, patient's QoL and expectations, as well as treatment costs have to be considered in the choice of treatment. However, to elucidate mechanisms of resistance, biomarkers of response and to better define personalized strategies in the treatment of cutaneous melanoma, larger clinical trials comparing combined versus sequential therapies are necessary.
Collapse
Affiliation(s)
- Regina Krattinger
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Joëlle Dornbierer
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich , Zurich, Switzerland
| |
Collapse
|
102
|
Klemen ND, Wang M, Rubinstein JC, Olino K, Clune J, Ariyan S, Cha C, Weiss SA, Kluger HM, Sznol M. Survival after checkpoint inhibitors for metastatic acral, mucosal and uveal melanoma. J Immunother Cancer 2020; 8:e000341. [PMID: 32209601 PMCID: PMC7103823 DOI: 10.1136/jitc-2019-000341] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Checkpoint inhibitors (CPIs) are thought to be effective against cutaneous melanoma in part because of the large burden of somatic mutations (neoantigens) generated from exposure to ultraviolet radiation. However, rare melanoma subtypes arising from acral skin, mucosal surfaces, and the uveal tract are largely sun-shielded. Genomic studies show these sun-shielded melanomas have a paucity of neoantigens and unique biology; they are thought to be largely resistant to immunotherapy. It has not been definitively shown that CPI improves survival in metastatic sun-shielded melanoma. METHODS We reviewed a single institutional experience using antibodies against CTLA-4, PD-1 and/or PD-L1 to treat patients with metastatic melanoma. Primary tumor histology was categorized as cutaneous, unknown, acral, mucosal, or uveal. We studied demographic data, treatment characteristics, and overall survival (OS) after CPI. RESULTS We treated 428 patients with metastatic melanoma from 2007 to 2019. Primary tumors were cutaneous in 283 (66%), unknown in 55 (13%), acral in 22 (5%), mucosal in 38 (9%), and uveal in 30 (7%). Patients with metastatic disease from cutaneous primary tumors had median OS after CPI of 45 months compared with 17 months for acral (p=0.047), 18 months for mucosal (p=0.003), and 12 months for uveal (p<0.001). For all patients with sun-shielded melanoma (n=90), first treatment with anti-PD-1 or anti-PD-L1 was followed by a median OS of 9 months compared with 18 months after anti-CTLA-4 (p=0.010) and 20 months after combination therapy (p=0.003). There were 21 patients who achieved actual 3-year survival; 20 received both anti-CTLA-4 and anti-PD-1, either sequentially or in combination. Over 80% of 3-year survivors with progressive disease were treated with local therapy after CPI. CONCLUSIONS Long survival in patients with metastatic melanoma from acral, mucosal, and uveal primary tumors was associated with receipt of both anti-CTLA-4 and anti-PD-1 antibodies. Complete responses were rare, and local therapy was frequently employed to control disease progression. While sun-shielded melanomas exhibit worse outcomes after CPI than cutaneous melanomas, with an aggressive multidisciplinary approach, 5-year survival is still possible for 25%-32% of these patients.
Collapse
Affiliation(s)
| | - Melinda Wang
- Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jill C Rubinstein
- Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kelly Olino
- Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - James Clune
- Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Stephan Ariyan
- Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Charles Cha
- Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sarah A Weiss
- Medical Oncology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Harriet M Kluger
- Medical Oncology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mario Sznol
- Medical Oncology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
103
|
Betof Warner A, Postow MA. Another Victory for Immune Checkpoint Blockade in Melanoma: Adjuvant Ipilimumab Over Interferon. J Clin Oncol 2020; 38:529-531. [DOI: 10.1200/jco.19.02988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Allison Betof Warner
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Michael A. Postow
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
104
|
Monti M, Consoli F, Vescovi R, Bugatti M, Vermi W. Human Plasmacytoid Dendritic Cells and Cutaneous Melanoma. Cells 2020; 9:E417. [PMID: 32054102 PMCID: PMC7072514 DOI: 10.3390/cells9020417] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/05/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
The prognosis of metastatic melanoma (MM) patients has remained poor for a long time. However, the recent introduction of effective target therapies (BRAF and MEK inhibitors for BRAFV600-mutated MM) and immunotherapies (anti-CTLA-4 and anti-PD-1) has significantly improved the survival of MM patients. Notably, all these responses are highly dependent on the fitness of the host immune system, including the innate compartment. Among immune cells involved in cancer immunity, properly activated plasmacytoid dendritic cells (pDCs) exert an important role, bridging the innate and adaptive immune responses and directly eliminating cancer cells. A distinctive feature of pDCs is the production of high amount of type I Interferon (I-IFN), through the Toll-like receptor (TLR) 7 and 9 signaling pathway activation. However, published data indicate that melanoma-associated escape mechanisms are in place to hijack pDC functions. We have recently reported that pDC recruitment is recurrent in the early phases of melanoma, but the entire pDC compartment collapses over melanoma progression. Here, we summarize recent advances on pDC biology and function within the context of melanoma immunity.
Collapse
Affiliation(s)
- Matilde Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Francesca Consoli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology, University of Brescia at ASST-Spedali Civili, 25123 Brescia, Italy;
| | - Raffaella Vescovi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - Mattia Bugatti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
| | - William Vermi
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.M.); (R.V.); (M.B.)
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
105
|
Almutairi AR, McBride A, Slack M, Erstad BL, Abraham I. Potential Immune-Related Adverse Events Associated With Monotherapy and Combination Therapy of Ipilimumab, Nivolumab, and Pembrolizumab for Advanced Melanoma: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:91. [PMID: 32117745 PMCID: PMC7033582 DOI: 10.3389/fonc.2020.00091] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/17/2020] [Indexed: 12/19/2022] Open
Abstract
Background: The use of ipilimumab, nivolumab, and pembrolizumab as monotherapies or in combination has transformed the management of advanced melanoma even though these drugs are associated with a new profile of immune-related adverse events (irAEs). The incidence of irAEs from clinical trials of these agents is an important factor for clinicians when treating patients with advanced melanoma. In the current study, we aimed to profile the incidence of potential irAEs of these agents when used as monotherapy and as combination therapy. Methods: We searched the Medline, Embase, and Cochrane databases; clinicaltrials.gov; and websites of regulatory agencies in the USA, Europe, Australia, and Japan for phase 1-3 trials of ipilimumab, nivolumab, and pembrolizumab for advanced melanoma. Random effect meta-analysis was utilized to profile the incidence of potential irAEs. Results: A total of 58 reports of 35 trials including 6,331 patients with advanced melanoma and reporting irAE data were included in the meta-analyses. We found higher incidences of potential irAEs in combination therapies vs. monotherapies for most of the types of irAEs. Among the monotherapies, ipilimumab users had the most frequent incidence of potential irAEs related to the gastrointestinal system (diarrhea, 29%; and colitis, 8%) and skin (rash, 31%; pruritus, 27%; and dermatitis, 10%), with hypophysitis in 4% of the patients. The most frequent potential irAEs among nivolumab users were maculopapular rash (13%), erythema (4%), hepatitis (3%), and infusion-related reactions (3%), while they were arthralgia (12%), hypothyroidism (8%), and hyperglycemia (6%), among pembrolizumab users. Conclusion: Especially the combination therapies tend to elevate the incidence of potential irAEs. Clinicians should be vigilant about irAEs following combination therapy as well as gastrointestinal and skin irAEs following ipilimumab therapy, in addition to being aware of potential irAEs leading to hyperglycemia, thyroid, hepatic, and musculoskeletal disorders following nivolumab and pembrolizumab therapy.
Collapse
Affiliation(s)
- Abdulaali R Almutairi
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Ali McBride
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| | - Marion Slack
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Brian L Erstad
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| | - Ivo Abraham
- Center for Health Outcomes and PharmacoEconomic Research, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,Department of Pharmacy Practice and Science, College of Pharmacy, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| |
Collapse
|
106
|
Gellrich FF, Schmitz M, Beissert S, Meier F. Anti-PD-1 and Novel Combinations in the Treatment of Melanoma-An Update. J Clin Med 2020; 9:E223. [PMID: 31947592 PMCID: PMC7019511 DOI: 10.3390/jcm9010223] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 12/15/2022] Open
Abstract
Until recently, distant metastatic melanoma was considered refractory to systemic therapy. A better understanding of the interactions between tumors and the immune system and the mechanisms of regulation of T-cells led to the development of immune checkpoint inhibitors. This review summarizes the current novel data on the treatment of metastatic melanoma with anti-programmed cell death protein 1 (PD-1) antibodies and anti-PD-1-based combination regimens, including clinical trials presented at major conference meetings. Immune checkpoint inhibitors, in particular anti-PD-1 antibodies such as pembrolizumab and nivolumab and the combination of nivolumab with the anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibody ipilimumab can achieve long-term survival for patients with metastatic melanoma. The anti-PD-1 antibodies nivolumab and pembrolizumab were also approved for adjuvant treatment of patients with resected metastatic melanoma. Anti-PD-1 antibodies appear to be well tolerated, and toxicity is manageable. Nivolumab combined with ipilimumab achieves a 5 year survival rate of more than 50% but at a cost of high toxicity. Ongoing clinical trials investigate novel immunotherapy combinations and strategies (e.g., Talimogene laherparepvec (T-VEC), Bempegaldesleukin (BEMPEG), incorporation or sequencing of targeted therapy, incorporation or sequencing of radiotherapy), and focus on poor prognosis groups (e.g., high tumor burden/LDH levels, anti-PD-1 refractory melanoma, and brain metastases).
Collapse
Affiliation(s)
- Frank Friedrich Gellrich
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (S.B.)
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, 01307 Dresden, Germany
| | - Marc Schmitz
- Institute of Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany;
- National Centre for Tumor Diseases, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Stefan Beissert
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (S.B.)
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, 01307 Dresden, Germany
| | - Friedegund Meier
- Department of Dermatology, University Hospital Carl Gustav Carus, TU Dresden, 01307 Dresden, Germany; (S.B.)
- Skin Cancer Center at the University Cancer Centre Dresden and National Center for Tumor Diseases, 01307 Dresden, Germany
| |
Collapse
|
107
|
Jardim DL, De Melo Gagliato D, Nikanjam M, Barkauskas DA, Kurzrock R. Efficacy and safety of anticancer drug combinations: a meta-analysis of randomized trials with a focus on immunotherapeutics and gene-targeted compounds. Oncoimmunology 2020; 9:1710052. [PMID: 32002305 PMCID: PMC6959453 DOI: 10.1080/2162402x.2019.1710052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/17/2019] [Accepted: 11/29/2019] [Indexed: 10/27/2022] Open
Abstract
Hundreds of trials are being conducted to evaluate combination of newer targeted drugs as well as immunotherapy. Our aim was to compare efficacy and safety of combination versus single non-cytotoxic anticancer agents. We searched PubMed (01/01/2001 to 03/06/2018) (and, for immunotherapy, ASCO and ESMO abstracts (2016 through March 2018)) for randomized clinical trials that compared a single non-cytotoxic agent (targeted, hormonal, or immunotherapy) versus a combination with another non-cytotoxic partner. Efficacy and safety endpoints were evaluated in a meta-analysis using a linear mixed-effects model (guidelines per PRISMA Report).We included 95 randomized comparisons (single vs. combination non-cytotoxic therapies) (59.4%, phase II; 41.6%, phase III trials) (29,175 patients (solid tumors)). Combinations most frequently included a hormonal agent and a targeted small molecule (23%). Compared to single non-cytotoxic agents, adding another non-cytotoxic drug increased response rate (odds ratio [OR]=1.61, 95%CI 1.40-1.84)and prolonged progression-free survival (hazard ratio [HR]=0.75, 95%CI 0.69-0.81)and overall survival (HR=0.87, 95%CI 0.81-0.94) (all p<0.001), which was most pronounced for the association between immunotherapy combinations and longer survival. Combinations also significantlyincreased the risk of high-grade toxicities (OR=2.42, 95%CI 1.98-2.97) (most notably for immunotherapy and small molecule inhibitors) and mortality at least possibly therapy related (OR: 1.33, 95%CI 1.15-1.53) (both p<0.001) (absolute mortality = 0.90% (single agent) versus 1.31% (combinations)) compared to single agents. In conclusion, combinations of non-cytotoxic drugs versus monotherapy in randomized cancer clinical trials attenuated safety, but increased efficacy, with the balance tilting in favor of combination therapy, based on the prolongation in survival.
Collapse
Affiliation(s)
- Denis L Jardim
- Department of Medical Oncology, Centro de Oncologia Hospital Sírio Libanês, São Paulo, Brazil
| | | | - Mina Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California, San Diego, CA, USA
| | - Donald A Barkauskas
- Biostatistics Division, Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California, San Diego, CA, USA
| |
Collapse
|
108
|
Abstract
While melanoma is less common than some other skin cancers, it is responsible for nearly 10,000 deaths in the USA each year alone. For many decades, very limited treatment options were available for patients with metastatic melanoma. However, recent breakthroughs have brought new hopes for patients and providers. While targeted therapy with BRAF and MEK inhibitors represents an important cornerstone in the treatment of metastatic melanoma, this chapter carefully reviews the past and current therapy options available, with a significant focus on immunotherapy-based approaches. In addition, we provide an overview of the results of recent advances in the adjuvant setting for patients with resected stage III and stage IV melanoma, as well as in patients with melanoma brain metastases. Finally, we provide a quick overview over the current research efforts in the field of immuno-oncology and melanoma.
Collapse
|
109
|
Rapid Affinity Maturation of Novel Anti-PD-L1 Antibodies by a Fast Drop of the Antigen Concentration and FACS Selection of Yeast Libraries. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6051870. [PMID: 31976323 PMCID: PMC6959147 DOI: 10.1155/2019/6051870] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 09/05/2019] [Indexed: 12/31/2022]
Abstract
The affinity engineering is a key step to increase the efficacy of therapeutic monoclonal antibodies and yeast surface display is the most widely used and powerful affinity maturation approach, achieving picomolar binding affinities. In this study, we provide an optimization of the yeast surface display methodology, applied to the generation of potentially therapeutic high affinity antibodies targeting the immune checkpoint PD-L1. In this approach, we coupled a 10-cycle error-prone mutagenesis of heavy chain complementarity determining region 3 of an anti‐PD-L1 scFv, previously identified by phage display, with high-throughput sequencing, to generate scFv-yeast libraries with high mutant frequency and diversity. In addition, we set up a novel, faster and effective selection scheme by fluorescence-activated cell sorting, based on a fast drop of the antigen concentration between the first and the last selection cycles, unlike the gradual decrease typical of current selection protocols. In this way we isolated 6 enriched mutated scFv-yeast clones overall, showing an affinity improvement for soluble PD-L1 protein compared to the parental scFv. As a proof of the potency of the novel approach, we confirmed that the antibodies converted from all the mutated scFvs retained the affinity improvement. Remarkably, the best PD-L1 binder among them also bound with a higher affinity to PD-L1 expressed in its native conformation on human-activated lymphocytes, and it was able to stimulate lymphocyte proliferation in vitro more efficiently than its parental antibody. This optimized technology, besides the identification of a new potential checkpoint inhibitor, provides a tool for the quick isolation of high affinity binders.
Collapse
|
110
|
Gorbet MJ, Ranjan A. Cancer immunotherapy with immunoadjuvants, nanoparticles, and checkpoint inhibitors: Recent progress and challenges in treatment and tracking response to immunotherapy. Pharmacol Ther 2019; 207:107456. [PMID: 31863820 DOI: 10.1016/j.pharmthera.2019.107456] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Chemotherapy, surgery, and radiation are accepted as the preferred treatment modalities against cancer, but in recent years the use of immunotherapeutic approaches has gained prominence as the fourth treatment modality in cancer patients. In this approach, a patient's innate and adaptive immune systems are activated to achieve clearance of occult cancerous cells. In this review, we discuss the preclinical and clinical immunotherapeutic (e.g., immunoadjuvants (in-situ vaccines, oncolytic viruses, CXC antagonists, device activated agents), organic and inorganic nanoparticles, and checkpoint blockade) that are under investigation for cancer therapy and diagnostics. Additionally, the innovations in imaging of immune cells for tracking therapeutic responses and limitations (e.g., toxicity, inefficient immunomodulation, etc.) are described. Existing data suggest that if immune therapy is optimized, it can be a real and potentially paradigm-shifting cancer treatment frontier.
Collapse
Affiliation(s)
- Michael-Joseph Gorbet
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74074, USA
| | - Ashish Ranjan
- Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74074, USA.
| |
Collapse
|
111
|
Extraocular Muscle Enlargement and Thyroid Eye Disease-like Orbital Inflammation Associated with Immune Checkpoint Inhibitor Therapy in Cancer Patients. Ophthalmic Plast Reconstr Surg 2019; 35:50-52. [PMID: 29927883 DOI: 10.1097/iop.0000000000001161] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
PURPOSE To describe thyroid eye disease (TED)-like orbital inflammatory syndrome in 3 cancer patients treated with immune checkpoint inhibitors. METHODS All consecutive patients treated by the senior author who were receiving immune checkpoint inhibitors and developed TED-like orbital inflammation were included. RESULTS Three cancer patients treated with immune checkpoint inhibitors developed orbital inflammation. The first patient was treated with a combination of a cytotoxic T-lymphocyte antigen-4 inhibitor and a programmed cell death protein 1 inhibitor and developed TED-like orbital inflammation with normal thyroid function and antibody levels. The second patient had a previous diagnosis of Graves disease without TED, and developed TED soon after initiating treatment with a programmed cell death protein 1 inhibitor. The third patient developed acute hyperthyroidism with symptomatic TED following treatment with an investigational cytotoxic T-lymphocyte antigen-4 inhibitor agent. All 3 patients were managed with either systemic steroids or observation, with resolution of their symptoms and without the need to halt immune checkpoint inhibitor treatment for their cancer. DISCUSSION AND CONCLUSIONS TED-like orbital inflammation may occur as a side effect of immune checkpoint inhibitor therapy with anti-cytotoxic T-lymphocyte antigen-4 or anti-PD-1 inhibitors. To the best of their knowledge, this is the first reported case of TED as a result of programmed cell death protein 1 inhibitor monotherapy. All 3 patients were treated with systemic steroids and responded quickly while continuing treatment with immune checkpoint inhibitors for their cancer. With increasing use of this class of drugs, clinicians should be familiar with the clinical manifestations and treatments for this adverse reaction.
Collapse
|
112
|
Goey AK, With MD, Agema BC, Hoop EOD, Singh RK, van der Veldt AA, Mathijssen RH, van Schaik RH, Bins S. Effects of pharmacogenetic variants on vemurafenib-related toxicities in patients with melanoma. Pharmacogenomics 2019; 20:1283-1290. [PMID: 31829834 DOI: 10.2217/pgs-2019-0101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The pharmacokinetics and pharmacodynamics of vemurafenib are characterized by a wide interpatient variability. Since multiple polymorphic enzymes and drug transporters are involved in vemurafenib pharmacokinetics, we studied associations of polymorphisms on vemurafenib-associated toxicities. Patients & methods: Prospectively collected samples of 97 melanoma patients treated with vemurafenib alone (n = 62) or in combination with cobimetinib (n = 35) were genotyped for ABCB1 (3435C>T), ABCG2 (421C>A, 34G>A) and CYP3A4 (*22, 15389C>T) polymorphisms. Associations between these variants and the incidence of toxicities were studied. Results: CYP3A4*22 was significantly associated with increased risk for grade ≥3 nausea, grade 1-4 hyperbilirubinemia, and cutaneous squamous cell carcinoma. ABCB1 3435C>T was a predictor for grade ≥3 toxicity. Conclusion: Genetic variants in CYP3A4 and ABCB1 are associated with vemurafenib-associated toxicities.
Collapse
Affiliation(s)
- Andrew Kl Goey
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Mirjam de With
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands.,Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bram C Agema
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Esther Oomen-De Hoop
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Rajbir K Singh
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Astrid Am van der Veldt
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands.,Department of Radiology & Nuclear Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ron Hj Mathijssen
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| | - Ron Hn van Schaik
- Department of Clinical Chemistry, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus Medical Center Cancer Institute, Rotterdam, The Netherlands
| |
Collapse
|
113
|
Lu J, Li L, Lan Y, Liang Y, Meng H. Immune checkpoint inhibitor-associated pituitary-adrenal dysfunction: A systematic review and meta-analysis. Cancer Med 2019; 8:7503-7515. [PMID: 31679184 PMCID: PMC6912062 DOI: 10.1002/cam4.2661] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/25/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
With the growing use of immune checkpoint inhibitors (ICIs), case reports of rare yet life-threatening pituitary-adrenal dysfunctions, particularly for hypopituitarism, are increasingly being published. In this analysis, we focus on these events by including the most recent publications and reports from early phase I/II and phase III clinical trials and comparing the incidence and risks across different ICI regimens. PubMed, Embase, and the Cochrane Library were systematically searched from inception to April 2019 for clinical trials that reported on pituitary-adrenal dysfunction. The rates of events, odds ratios (ORs), and 95% confidence intervals (CIs) were obtained using random effects meta-analysis. The analyses included data from 160 trials involving 40 432 participants. The rate was 2.43% (95% CI, 1.73%-3.22%) for all-grade adrenal insufficiency and 3.25% (95% CI, 2.15%-4.51%) for hypophysitis. Compared with the placebo or other therapeutic regimens, ICI agents were associated with a higher incidence of serious-grade adrenal insufficiency (OR 3.19, 95% CI, 1.84 to 5.54) and hypophysitis (OR 4.77, 95% CI, 2.60 to 8.78). Among 71 serious-grade hypopituitarism instances in 12 336 patients, there was a significant association between ICIs and hypopituitarism (OR 3.62, 95% CI, 1.86 to 7.03). Substantial heterogeneity was noted across the studies for the rates of these events, which in part was attributable to the different types of ICIs and varied phases of the clinical trials. Although the rates of these events were low, the risk was increased following ICI-based treatment, particularly for CTLA-4 inhibitors, which were associated with a higher incidence of pituitary-adrenal dysfunction than PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Jingli Lu
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Lulu Li
- Department of PharmacyWuhan No.1 HospitalWuhanHubeiChina
| | - Yan Lan
- Department of PharmacyHuangshi Center HospitalHuangshiHubeiChina
| | - Yan Liang
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| | - Haiyang Meng
- Department of PharmacyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Henan Key Laboratory of Precision Clinical PharmacyZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
114
|
Xu H, Tan P, Ai J, Zhang S, Zheng X, Liao X, Yang L, Wei Q. Antitumor Activity and Treatment-Related Toxicity Associated With Nivolumab Plus Ipilimumab in Advanced Malignancies: A Systematic Review and Meta-Analysis. Front Pharmacol 2019; 10:1300. [PMID: 31749704 PMCID: PMC6844121 DOI: 10.3389/fphar.2019.01300] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 10/11/2019] [Indexed: 02/05/2023] Open
Abstract
Combining immune checkpoint inhibitors has shown its efficacy compared to monotherapy in advanced malignancies. We conducted this meta-analysis to provide latest evidence on the objective response rate (ORR) and incidence of treatment-related high-grade adverse events (AEs) during nivolumab and ipilimumab combination treatment and further explore from different drug dose level. PubMed and the 2019 American Society of Clinical Oncology (ASCO) annual meeting abstracts were searched for qualified clinical trials up to June 2019. Of the 23 clinical trials (13 from publications and 11 from ASCO abstracts) included, 2,114 and 2,674 patients were eligible for efficacy and safety analysis, respectively. Pooled analysis suggested that the overall ORR was achieved in 34.5% [95% confidence interval (CI), 29.1-40.4%] of patients. There was no significant difference between nivolumab 3 mg/kg plus ipilimumab 1 mg/kg every 3 weeks (N3I1-Q3W) and nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks (N1I3-Q3W) arms in ORR [30.8% vs 41%; odds ratio (OR), 0.72; 95% CI, 0.39-1.30; P = 0.275]. Grade 3-4 AEs related to combination therapy occurred in 39.9% (95% CI, 33.5-46.7%) of patients; the most commonly reported grade 3-4 treatment-related AEs were diarrhea (5.28%), colitis (3.96%) and increased alanine aminotransferase (3.51%). Incidence of grade 3-4 AEs were significant lower in N3I1-Q3W arm than in N1I3-Q3W arm (31.3% vs 55.9%; OR 0.52; 95% CI, 0.32-0.87; P = 0.012). Treatment-related death was rare and occurred in 2.0% (95% CI, 1.5-2.7%) of patients. Our comprehensive study provides more precise data on the incidence of treatment-related high-grade AEs and ORR among patients receiving nivolumab and ipilimumab combination regimens. Patients on the N3I1-Q3W arm had comparable ORR and significantly occurred less grade 3-4 AEs than patients on the N1I3-Q3W arm. Our finding is of great importance in assisting clinical trial design and clinical medication choice.
Collapse
Affiliation(s)
- Hang Xu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Ping Tan
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyu Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaonan Zheng
- West China Medical School, Sichuan University, Chengdu, China
| | - Xinyang Liao
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Yang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
115
|
Eggermont AMM. The impact of the immunotherapy revolution on lymph nodal surgery. Bull Cancer 2019; 107:640-641. [PMID: 31610910 DOI: 10.1016/j.bulcan.2019.05.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/12/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Alexander M M Eggermont
- University Paris-Sud, Gustave Roussy Cancer Campus Grand Paris, 114, rue Edouard-Vaillant, 94800 Villejuif, France.
| |
Collapse
|
116
|
Pires da Silva I, Lo S, Quek C, Gonzalez M, Carlino MS, Long GV, Menzies AM. Site‐specific response patterns, pseudoprogression, and acquired resistance in patients with melanoma treated with ipilimumab combined with anti–PD‐1 therapy. Cancer 2019; 126:86-97. [DOI: 10.1002/cncr.32522] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/15/2019] [Accepted: 08/09/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Ines Pires da Silva
- Melanoma Institute Australia Sydney New South Wales Australia
- Central Clinical School The University of Sydney Sydney New South Wales Australia
| | - Serigne Lo
- Melanoma Institute Australia Sydney New South Wales Australia
- Central Clinical School The University of Sydney Sydney New South Wales Australia
| | - Camelia Quek
- Melanoma Institute Australia Sydney New South Wales Australia
- Central Clinical School The University of Sydney Sydney New South Wales Australia
| | - Maria Gonzalez
- Melanoma Institute Australia Sydney New South Wales Australia
| | - Matteo S. Carlino
- Melanoma Institute Australia Sydney New South Wales Australia
- Western Clinical School The University of Sydney Sydney New South Wales Australia
- Department of Medical Oncology Westmead and Blacktown Hospitals Sydney New South Wales Australia
| | - Georgina V. Long
- Melanoma Institute Australia Sydney New South Wales Australia
- Department of Medical Oncology Royal North Shore and Mater Hospitals Sydney New South Wales Australia
- Northern Clinical School The University of Sydney Sydney New South Wales Australia
| | - Alexander M. Menzies
- Melanoma Institute Australia Sydney New South Wales Australia
- Department of Medical Oncology Royal North Shore and Mater Hospitals Sydney New South Wales Australia
- Northern Clinical School The University of Sydney Sydney New South Wales Australia
| |
Collapse
|
117
|
Myocarditis following the use of different immune checkpoint inhibitor regimens: A real-world analysis of post-marketing surveillance data. Int Immunopharmacol 2019; 76:105866. [PMID: 31491729 DOI: 10.1016/j.intimp.2019.105866] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/20/2019] [Accepted: 08/26/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUNDS Although myocarditis has been reported in patients treated with immune checkpoint inhibitors (ICIs), there are few real-world studies to compare the occurrences and characteristics of myocarditis after different ICI regimens. METHODS Disproportionality analysis and Bayesian analysis were utilized for data mining of the suspected adverse events of myocarditis after ICIs use based on the Food and Drug Administration's Adverse Event Reporting System (FAERS) from January 2004 to June 2018. The times to onset and fatality rates of myocarditis following different ICI regimens were also compared. RESULTS A total of 315 reports of myocarditis adverse events were identified with ICIs. Among 6 ICI monotherapies, avelumab had the highest association with myocarditis based on the highest reporting odds ratio (ROR = 42.65, 95% two-sided CI = 15.86-114.72), proportional reporting ratio (PRR = 42.61, χ2 = 159.63) and empirical Bayes geometric mean (EBGM = 41.87, 95% one-sided CI = 15.57). The combination therapies of ipilimumab plus pembrolizumab or nivolumab had higher RORs, PRRs and EBGMs than did pembrolizumab or nivolumab monotherapy. Myocarditis associated with the ipilimumab plus nivolumab treatment appeared to have earlier onset (16.5 [IQR 14-29.75] days vs 32 [IQR 16-77] days, p<0.01) and higher fatality rate (65.75% vs 50.40%, p<0.05) than that associated with nivolumab monotherapy. CONCLUSIONS Analysis of FAERS data provides more precise profile on the occurrences and characteristics of myocarditis after different ICI regimens. The findings support continued surveillance, risk factor identification, and comparative studies.
Collapse
|
118
|
Abstract
Immune checkpoint blockers have revolutionized cancer treatment in recent years. These agents are now approved for the treatment of several malignancies, including melanoma, squamous and non-squamous non-small cell lung cancer, renal cell carcinoma, urothelial carcinoma, and head and neck squamous cell carcinoma. Studies have demonstrated the significant impact of immunotherapy versus standard of care on patient outcomes, including durable response and extended survival. The use of immunotherapy-based combination therapy has been shown to further extend duration of response and survival. Immunotherapies function through modulation of the immune system, which can lead to immune-mediated adverse events (imAEs). These include a range of dermatologic, gastrointestinal, endocrine, and hepatic toxicities, as well as other less common inflammatory events. ImAEs are typically low grade and manageable when identified early and treated with appropriate measures. Identifying the right patient for the right therapy will become more important as new immunotherapies and immunotherapy-based combinations are approved and costs of cancer care continue to rise.
Collapse
|
119
|
Weiss SA, Wolchok JD, Sznol M. Immunotherapy of Melanoma: Facts and Hopes. Clin Cancer Res 2019; 25:5191-5201. [PMID: 30923036 PMCID: PMC6726509 DOI: 10.1158/1078-0432.ccr-18-1550] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/21/2019] [Accepted: 03/25/2019] [Indexed: 12/14/2022]
Abstract
Melanoma is among the most sensitive of malignancies to immune modulation. Although multiple trials conducted over decades with vaccines, cytokines, and cell therapies demonstrated meaningful responses in a small subset of patients with metastatic disease, a true increase in overall survival (OS) within a randomized phase III trial was not observed until the development of anti-CTLA-4 (ipilimumab). Further improvements in OS for metastatic disease were observed with the anti-PD-1-based therapies (nivolumab, pembrolizumab) as single agents or combined with ipilimumab. A lower bound for expected 5-year survival for metastatic melanoma is currently approximately 35% and could be as high as 50% for the nivolumab/ipilimumab combination among patients who would meet criteria for clinical trials. Moreover, a substantial fraction of long-term survivors will likely remain progression-free without continued treatment. The hope and major challenge for the future is to understand the immunobiology of tumors with primary or acquired resistance to anti-PD-1 or anti-PD-1/anti-CTLA-4 and to develop effective immune therapies tailored to individual patient subsets not achieving long-term clinical benefit. Additional goals include optimal integration of immune therapy with nonimmune therapies, the development and validation of predictive biomarkers in the metastatic setting, improved prognostic and predictive biomarkers for the adjuvant setting, understanding mechanisms of and decreasing toxicity, and optimizing the duration of therapy.
Collapse
Affiliation(s)
- Sarah A Weiss
- Yale University School of Medicine, New Haven, Connecticut.
| | - Jedd D Wolchok
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York
| | - Mario Sznol
- Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
120
|
Queirolo P, Boutros A, Tanda E, Spagnolo F, Quaglino P. Immune-checkpoint inhibitors for the treatment of metastatic melanoma: a model of cancer immunotherapy. Semin Cancer Biol 2019; 59:290-297. [PMID: 31430555 DOI: 10.1016/j.semcancer.2019.08.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/23/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022]
Abstract
Melanoma has always been described as an immunogenic tumor. Despite that, until 2011 the standard of care in metastatic melanoma was chemotherapy, with low response rates and no clear impact on overall survival. Melanoma was the first cancer type to drive the use of immune-checkpoint inhibitors into clinical practice, which revolutionized the therapeutic paradigm not only in melanoma, but also in an increasing number of tumors. In this review, the preclinical bases and the main clinical studies that led to the approval of immune-checkpoint inhibitors in advanced melanoma will be described with insights on novel combinations of treatments and on prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Paola Queirolo
- IRCCS Ospedale Policlinico San Martino, Skin Cancer Unit, Genoa, Italy
| | - Andrea Boutros
- IRCCS Ospedale Policlinico San Martino, Skin Cancer Unit, Genoa, Italy
| | - Enrica Tanda
- IRCCS Ospedale Policlinico San Martino, Skin Cancer Unit, Genoa, Italy
| | | | - Pietro Quaglino
- Dermatologic Clinic, Department of Medical Sciences, University of Turin Medical School, Italy
| |
Collapse
|
121
|
Abstract
Immunotherapy has dramatically improved the prognosis for patients with melanoma and has become the cornerstone of treatment for those with advanced disease. The role of immunotherapy continues to expand with multiple new agents approved in the adjuvant as well as metastatic setting, as first-line therapy and beyond. We review the currently approved drugs for the treatment of melanoma, along with clinical trial data, adverse side effects, response assessment and future directions.
Collapse
Affiliation(s)
- Emily Feld
- UDepartment of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- UDepartment of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
122
|
Vanella V, Festino L, Trojaniello C, Vitale MG, Sorrentino A, Paone M, Ascierto PA. The Role of BRAF-Targeted Therapy for Advanced Melanoma in the Immunotherapy Era. Curr Oncol Rep 2019; 21:76. [DOI: 10.1007/s11912-019-0827-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
123
|
Wan X, Luo X, Tan C, Zeng X, Zhang Y, Peng L. First‐line atezolizumab in addition to bevacizumab plus chemotherapy for metastatic, nonsquamous non–small cell lung cancer: A United States–based cost‐effectiveness analysis. Cancer 2019; 125:3526-3534. [PMID: 31287562 DOI: 10.1002/cncr.32368] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/10/2019] [Accepted: 06/06/2019] [Indexed: 12/25/2022]
Affiliation(s)
- XiaoMin Wan
- Department of Pharmacy The Second Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacy Central South University Changsha Hunan China
| | - Xia Luo
- Department of Pharmacy The Second Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacy Central South University Changsha Hunan China
| | - ChongQing Tan
- Department of Pharmacy The Second Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacy Central South University Changsha Hunan China
| | - XiaoHui Zeng
- The PET‐CT Center The Second Xiangya Hospital, Central South University Changsha Hunan China
| | - YuCong Zhang
- Department of Pharmacy The Second Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacy Central South University Changsha Hunan China
| | - LiuBao Peng
- Department of Pharmacy The Second Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacy Central South University Changsha Hunan China
| |
Collapse
|
124
|
Kennedy LB, Salama AKS. A Review of Immune-Mediated Adverse Events in Melanoma. Oncol Ther 2019; 7:101-120. [PMID: 32699983 PMCID: PMC7359990 DOI: 10.1007/s40487-019-0096-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Indexed: 12/16/2022] Open
Abstract
The use of checkpoint inhibitor-based immunotherapy has transformed the treatment landscape for melanoma as well as many other cancer types. With the ability to potentiate tumor-specific immune responses, these agents can result in durable tumor control. However, this activation of the immune system can lead to a unique constellation of side effects, distinct from other cancer therapies, collectively termed immune-mediated adverse events (irAEs). This review will focus on irAEs and guidelines for management related to the most clinically relevant checkpoint inhibitors, those that target programmed death receptor-1 (PD-1) and cytotoxic T lymphocyte antigen-4 (CTLA-4).
Collapse
|
125
|
Survival and prognosis of individuals receiving programmed cell death 1 inhibitor with and without immunologic cutaneous adverse events. J Am Acad Dermatol 2019; 82:311-316. [PMID: 31233857 DOI: 10.1016/j.jaad.2019.06.035] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 05/05/2019] [Accepted: 06/17/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND The treatment response to new immunotherapy in advanced melanoma patients remains varied between individuals. Immune-related cutaneous side effects might have prognostic value. OBJECTIVE To determine whether development of ≥1 of the 3 immune-mediated cutaneous events (eczema, lichenoid reaction, or vitiligo-like depigmentation) is associated with improved progression-free survival. METHODS A cohort study of adults with stage IIIC-IV melanoma treated with pembrolizumab or nivolumab during May 1, 2012-February 1, 2018, at Westmead Hospital, Sydney, Australia. Treatment response was based on iRECIST version 1.1. RESULTS In total, 82 patients of an average age of 59.9 years were included. Median follow-up was 40.7 months; 33 patients had ≥1 target skin reaction. Skin reactions developed in one-third of individuals by 6 months. At any given time, the instantaneous risk of disease progression and death was lower for individuals who had ≥1 cutaneous adverse event (CAE) develop. Compared with individuals with no CAE, the hazard ratio for disease progression and death for individuals who had ≥1 CAE develop was 0.46 (95% confidence interval 0.23-0.91; P = .025) by the time-dependent Cox proportional hazards model. LIMITATIONS Single-center study. CONCLUSION This study demonstrates an association between the development of ≥1 of 3 CAEs and improved progression-free survival in this cohort of patients.
Collapse
|
126
|
Long GV, Dummer R, Hamid O, Gajewski TF, Caglevic C, Dalle S, Arance A, Carlino MS, Grob JJ, Kim TM, Demidov L, Robert C, Larkin J, Anderson JR, Maleski J, Jones M, Diede SJ, Mitchell TC. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol 2019; 20:1083-1097. [PMID: 31221619 DOI: 10.1016/s1470-2045(19)30274-8] [Citation(s) in RCA: 670] [Impact Index Per Article: 111.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND Immunotherapy combination treatments can improve patient outcomes. Epacadostat, an IDO1 selective inhibitor, and pembrolizumab, a PD-1 inhibitor, showed promising antitumour activity in the phase 1-2 ECHO-202/KEYNOTE-037 study in advanced melanoma. In this trial, we aimed to compare progression-free survival and overall survival in patients with unresectable stage III or IV melanoma receiving epacadostat plus pembrolizumab versus placebo plus pembrolizumab. METHODS In this international, randomised, placebo-controlled, double-blind, parallel-group, phase 3 trial, eligible participants were aged 18 years or older, with unresectable stage III or IV melanoma previously untreated with PD-1 or PD-L1 checkpoint inhibitors, an ECOG performance status of 0 or 1, and had a known BRAFV600 mutant status or consented to BRAFV600 mutation testing during screening. Patients were stratified by PD-L1 expression and BRAFV600 mutation status and randomly assigned (1:1) through a central interactive voice and integrated web response system to receive epacadostat 100 mg orally twice daily plus pembrolizumab 200 mg intravenously every 3 weeks or placebo plus pembrolizumab for up to 2 years. We used block randomisation with a block size of four in each stratum. Primary endpoints were progression-free survival and overall survival in the intention-to-treat population. The safety analysis population included randomly assigned patients who received at least one dose of study treatment. The study was stopped after the second interim analysis; follow-up for safety is ongoing. This study is registered with ClinicalTrials.gov, number NCT02752074. FINDINGS Between June 21, 2016, and Aug 7, 2017, 928 patients were screened and 706 patients were randomly assigned to receive epacadostat plus pembrolizumab (n=354) or placebo plus pembrolizumab (n=352). Median follow-up was 12·4 months (IQR 10·3-14·5). No significant differences were found between the treatment groups for progression-free survival (median 4·7 months, 95% CI 2·9-6·8, for epacadostat plus pembrolizumab vs 4·9 months, 2·9-6·8, for placebo plus pembrolizumab; hazard ratio [HR] 1·00, 95% CI 0·83-1·21; one-sided p=0·52) or overall survival (median not reached in either group; epacadostat plus pembrolizumab vs placebo plus pembrolizumab: HR 1·13, 0·86-1·49; one-sided p=0·81). The most common grade 3 or worse treatment-related adverse event was lipase increase, which occurred in 14 (4%) of 353 patients receiving epacadostat plus pembrolizumab and 11 (3%) of 352 patients receiving placebo plus pembrolizumab. Treatment-related serious adverse events were reported in 37 (10%) of 353 patients receiving epacadostat plus pembrolizumab and 32 (9%) of 352 patients receiving placebo plus pembrolizumab. There were no treatment-related deaths in either treatment group. INTERPRETATION Epacadostat 100 mg twice daily plus pembrolizumab did not improve progression-free survival or overall survival compared with placebo plus pembrolizumab in patients with unresectable or metastatic melanoma. The usefulness of IDO1 inhibition as a strategy to enhance anti-PD-1 therapy activity in cancer remains uncertain. FUNDING Incyte Corporation, in collaboration with Merck Sharp & Dohme.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, University of Sydney, Royal North Shore and Mater Hospitals, Sydney, NSW, Australia.
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zürich, Zurich, Switzerland
| | - Omid Hamid
- The Angeles Clinic and Research Institute, Los Angeles, CA, USA
| | - Thomas F Gajewski
- Department of Pathology, University of Chicago Medical Center, Chicago, IL, USA
| | - Christian Caglevic
- Oncology Department, Clinica Alemana Santiago, Universidad del Desarrollo, Santiago, Chile
| | - Stephane Dalle
- Hospices Civils De Lyon, Cancer Research Center of Lyon, Claude Bernard University Lyon, Pierre Benite, France
| | - Ana Arance
- Medical Oncology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Matteo S Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | - Jean-Jacques Grob
- Service de Dermatologie et Cancérologie Cutanée, Aix-Marseille University, Marseille, France
| | - Tae Min Kim
- Department of Haemato-Oncology, Seoul National University Hospital, Seoul, South Korea
| | - Lev Demidov
- N N Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Caroline Robert
- Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - James Larkin
- The Royal Marsden NHS Foundation Trust, London, UK
| | | | | | | | | | - Tara C Mitchell
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
127
|
Rotte A. Combination of CTLA-4 and PD-1 blockers for treatment of cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:255. [PMID: 31196207 PMCID: PMC6567914 DOI: 10.1186/s13046-019-1259-z] [Citation(s) in RCA: 656] [Impact Index Per Article: 109.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022]
Abstract
Targeting checkpoints of immune cell activation has been demonstrated to be the most effective approach for activation of anti-tumor immune responses. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1), both inhibitory checkpoints commonly seen on activated T-cells have been found to be the most reliable targets for the treatment of cancer. Six drugs targeting PD-1 or its ligand PD-L1 and one drug targeting CTLA-4 have been approved for treatment of different types of cancers and several others are in advanced stages of development. The drugs when administered as monotherapy had dramatic increase in durable response rates and had manageable safety profile, but more than 50% of patients failed to respond to treatment. Combination of CTLA-4 and PD-1 blockers was then evaluated to increase the response rates in patients, and ipilimumab (anti-CTLA-4) plus nivolumab (anti-PD-1) combination was shown to significantly enhance efficacy in metastatic melanoma patients. Subsequently, ipilimumab plus nivolumab was approved for treatment of metastatic melanoma, advanced renal cell carcinoma and metastatic colorectal cancer with MMR/MSI-H aberrations. The success of combination encouraged multiple clinical studies in other cancer types. Efficacy of the combination has been shown in a number of published studies and is under evaluation in multiple ongoing studies. This review aims to support future research in combination immunotherapy by discussing the basic details of CTLA-4 and PD-1 pathways and the results from clinical studies that evaluated combination of CTLA-4 and PD-1/PD-L1 blockers.
Collapse
Affiliation(s)
- Anand Rotte
- Clinical & Regulatory Affairs, Nevro Corp, 1800 Bridge Parkway, Redwood City, CA, 94065, USA.
| |
Collapse
|
128
|
Gu L, Khadaroo PA, Su H, Kong L, Chen L, Wang X, Li X, Zhu H, Zhong X, Pan J, Chen M. The safety and tolerability of combined immune checkpoint inhibitors (anti-PD-1/PD-L1 plus anti-CTLA-4): a systematic review and meta-analysis. BMC Cancer 2019; 19:559. [PMID: 31182049 PMCID: PMC6558837 DOI: 10.1186/s12885-019-5785-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/31/2019] [Indexed: 12/18/2022] Open
Abstract
Background The future of combined immunotherapy (a PD-1/PD-L1 plus a CTLA-4 antagonist) is very bright. However, besides improving efficacy, combined therapy increases treatment-related adverse events (TRAEs). Also, the clinical application is limited in some solid tumors. Methods This paper purports to investigate the TRAEs for the combined immunotherapy aiming for a more appropriate utilization of immune checkpoint inhibitors (ICIs) in clinical practice through a meta-analysis. Results A total of 17 eligible studies covering 2626 patients were selected for a meta-analysis based on specified inclusion and exclusion criteria. The incidence rates of any grade and grade 3 or higher TRAEs were 88% (95%CI, 84–92%) and 41% (95%CI, 35–47%), respectively. The overall incidence of any grade TRAEs leading to discontinuation of treatment was 20% (95%CI, 16–24%). The incidence rate of treatment related deaths was 4.3‰ (95%CI, 1.4‰-8.4‰). Analysis showed that NIVO1 + IPI3 cohort had higher incidences of grade 3 or higher TRAEs (RR = 1.77, 95%CI, 1.34–2.34, p < 0.0001) and any grade TRAEs leading to discontinuation of treatment (RR = 1.81, 95%CI, 1.08–3.04, P = 0.02), compared with NIVO3 + IPI1 regimen. Conclusions The combined therapy had high TRAEs. The TRAEs, especially grade 3 or higher, led to discontinuation of the treatment. Furthermore, the incidence of treatment-related deaths was rare. Moreover, the NIVO3 + IPI1 regimen, regardless of efficacy, is more recommended because of better tolerance and lower adverse events. Electronic supplementary material The online version of this article (10.1186/s12885-019-5785-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lihu Gu
- Department of General Surgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | | | - Hui Su
- Department of General Surgery, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Liya Kong
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Liangliang Chen
- Department of Surgical Oncology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Xianfa Wang
- Department of General Surgery, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, China
| | - Xinlong Li
- Department of General Surgery, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, China
| | - Hepan Zhu
- Department of General Surgery, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, China
| | - Xin Zhong
- Department of General Surgery, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, China
| | - Junhai Pan
- Department of General Surgery, Zhejiang University School of Medicine Sir Run Run Shaw Hospital, Hangzhou, Zhejiang, China
| | - Manman Chen
- Affiliated Hospital of Medical School Ningbo University and Ningbo City Third Hospital, No. 247, Renming Road, Ningbo, 315020, Zhejiang, China.
| |
Collapse
|
129
|
Rozeman EA, Menzies AM, van Akkooi ACJ, Adhikari C, Bierman C, van de Wiel BA, Scolyer RA, Krijgsman O, Sikorska K, Eriksson H, Broeks A, van Thienen JV, Guminski AD, Acosta AT, Ter Meulen S, Koenen AM, Bosch LJW, Shannon K, Pronk LM, Gonzalez M, Ch'ng S, Grijpink-Ongering LG, Stretch J, Heijmink S, van Tinteren H, Haanen JBAG, Nieweg OE, Klop WMC, Zuur CL, Saw RPM, van Houdt WJ, Peeper DS, Spillane AJ, Hansson J, Schumacher TN, Long GV, Blank CU. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol 2019; 20:948-960. [PMID: 31160251 DOI: 10.1016/s1470-2045(19)30151-2] [Citation(s) in RCA: 352] [Impact Index Per Article: 58.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND The outcome of patients with macroscopic stage III melanoma is poor. Neoadjuvant treatment with ipilimumab plus nivolumab at the standard dosing schedule induced pathological responses in a high proportion of patients in two small independent early-phase trials, and no patients with a pathological response have relapsed after a median follow up of 32 months. However, toxicity of the standard ipilimumab plus nivolumab dosing schedule was high, preventing its broader clinical use. The aim of the OpACIN-neo trial was to identify a dosing schedule of ipilimumab plus nivolumab that is less toxic but equally effective. METHODS OpACIN-neo is a multicentre, open-label, phase 2, randomised, controlled trial. Eligible patients were aged at least 18 years, had a WHO performance status of 0-1, had resectable stage III melanoma involving lymph nodes only, and measurable disease according to the Response Evaluation Criteria in Solid Tumors version 1.1. Patients were enrolled from three medical centres in Australia, Sweden, and the Netherlands, and were randomly assigned (1:1:1), stratified by site, to one of three neoadjuvant dosing schedules: group A, two cycles of ipilimumab 3 mg/kg plus nivolumab 1 mg/kg once every 3 weeks intravenously; group B, two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg once every 3 weeks intravenously; or group C, two cycles of ipilimumab 3 mg/kg once every 3 weeks directly followed by two cycles of nivolumab 3 mg/kg once every 2 weeks intravenously. The investigators, site staff, and patients were aware of the treatment assignment during the study participation. Pathologists were masked to treatment allocation and all other data. The primary endpoints were the proportion of patients with grade 3-4 immune-related toxicity within the first 12 weeks and the proportion of patients achieving a radiological objective response and pathological response at 6 weeks. Analyses were done in all patients who received at least one dose of study drug. This trial is registered with ClinicalTrials.gov, number NCT02977052, and is ongoing with an additional extension cohort and to complete survival analysis. FINDINGS Between Nov 24, 2016 and June 28, 2018, 105 patients were screened for eligibility, of whom 89 (85%) eligible patients were enrolled and randomly assigned to one of the three groups. Three patients were excluded after randomisation because they were found to be ineligible, and 86 received at least one dose of study drug; 30 patients in group A, 30 in group B, and 26 in group C (accrual to this group was closed early upon advice of the Data Safety Monitoring Board on June 4, 2018 because of severe adverse events). Within the first 12 weeks, grade 3-4 immune-related adverse events were observed in 12 (40%) of 30 patients in group A, six (20%) of 30 in group B, and 13 (50%) of 26 in group C. The difference in grade 3-4 toxicity between group B and A was -20% (95% CI -46 to 6; p=0·158) and between group C and group A was 10% (-20 to 40; p=0·591). The most common grade 3-4 adverse events were elevated liver enzymes in group A (six [20%)]) and colitis in group C (five [19%]); in group B, none of the grade 3-4 adverse events were seen in more than one patient. One patient (in group A) died 9·5 months after the start of treatment due to the consequences of late-onset immune-related encephalitis, which was possibly treatment-related. 19 (63% [95% CI 44-80]) of 30 patients in group A, 17 (57% [37-75]) of 30 in group B, and nine (35% [17-56]) of 26 in group C achieved a radiological objective response, while pathological responses occurred in 24 (80% [61-92]) patients in group A, 23 (77% [58-90]) in group B, and 17 (65% [44-83]) in group C. INTERPRETATION OpACIN-neo identified a tolerable neoadjuvant dosing schedule (group B: two cycles of ipilimumab 1 mg/kg plus nivolumab 3 mg/kg) that induces a pathological response in a high proportion of patients and might be suitable for broader clinical use. When more mature data confirm these early observations, this schedule should be tested in randomised phase 3 studies versus adjuvant therapies, which are the current standard-of-care systemic therapy for patients with stage III melanoma. FUNDING Bristol-Myers Squibb.
Collapse
Affiliation(s)
| | - Alexander M Menzies
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | - Chandra Adhikari
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | | | | | - Richard A Scolyer
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Hanna Eriksson
- Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | | | - Alexander D Guminski
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | | | | | | | | | - Kerwin Shannon
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Loes M Pronk
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Maria Gonzalez
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | - Sydney Ch'ng
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | - Jonathan Stretch
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Stijn Heijmink
- The Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | | | - Omgo E Nieweg
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Robyn P M Saw
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia; Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | | | | | - Andrew J Spillane
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia; Royal North Shore and Mater Hospitals, Sydney, NSW, Australia
| | - Johan Hansson
- Karolinska University Hospital and Karolinska Institute, Stockholm, Sweden
| | | | - Georgina V Long
- Melanoma Institute of Australia, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
130
|
Schvartsman G, Taranto P, Glitza IC, Agarwala SS, Atkins MB, Buzaid AC. Management of metastatic cutaneous melanoma: updates in clinical practice. Ther Adv Med Oncol 2019; 11:1758835919851663. [PMID: 31205512 PMCID: PMC6535734 DOI: 10.1177/1758835919851663] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
In recent years, several drugs have been approved for the treatment of patients with metastatic cutaneous melanoma, completely reshaping the landscape of this aggressive disease. Immune therapy with cytotoxic T-lymphocyte antigen 4 and programmed cell death-1 inhibitors yielded significant and durable responses, achieving long-term disease control in up to 40% of the patients. BRAF inhibitors (BRAFi), in combination with MEK inhibitors, also resulted in improved overall survival compared with single-agent BRAFi in patients with BRAFV600-mutated metastatic melanoma. The optimized sequencing and duration of treatment, however, is yet to be found. In this article, we thoroughly review current data and discuss how to best sequence the various treatment modalities available at present, based on four distinct clinical presentations commonly seen in clinic. In addition, we review treatment options beyond checkpoint inhibitors and targeted therapy, which may be required by patients failing such effective treatments.
Collapse
Affiliation(s)
- Gustavo Schvartsman
- Centro de Oncologia e Hematologia - Hospital Israelita Albert Einstein, 627 Albert Einstein Avenue, São Paulo, SP 05653-120, Brazil
| | - Patricia Taranto
- Department of Medical Oncology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Isabella C Glitza
- Department of Melanoma Medical Oncology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Sanjiv S Agarwala
- Department of Hematology and Oncology, and Temple University, Easton, PA, USA
| | - Michael B Atkins
- Department of Oncology, Georgetown University School of Medicine, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | - Antonio C Buzaid
- Department of Medical Oncology, Hospital Israelita Albert Einstein, São Paulo, SP, Brazila and Department of Medical Oncology, A Beneficência Portuguesa de São Paulo - BP, São Paulo, SP, Brazil
| |
Collapse
|
131
|
Pavlick AC, Fecher L, Ascierto PA, Sullivan RJ. Frontline Therapy for BRAF-Mutated Metastatic Melanoma: How Do You Choose, and Is There One Correct Answer? Am Soc Clin Oncol Educ Book 2019; 39:564-571. [PMID: 31099689 DOI: 10.1200/edbk_243071] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetic analysis of melanoma has allowed us to identify a population of patients who have more aggressive disease and harbor the driver mutation BRAF. This mutation is found in approximately 50% of metastatic disease and provides a target for focused therapies to control this disease. These responses are usually brisk; however, they lack the durability of immunotherapy. Frontline therapy for patients with BRAF-mutated melanoma is not as straightforward as prescribing BRAF/MEK inhibitors. Prior trials of combination immunotherapy demonstrate similar responses and durability of responses in patients with BRAF wild-type as well as BRAF-mutated disease. Decisions about immunotherapy, targeted therapy, or the combination of immunotherapy with targeted therapy require an oncologist to evaluate multiple factors to select which treatment option is best for the patient. Trials for metastatic melanoma have included biomarkers as secondary endpoints and aim to identify some way to predict a response, or lack thereof, to therapy. Here, we discuss the utility and reliability of biomarkers in determining therapy for patients with BRAF-mutated metastatic melanoma and discuss combination immunotherapy with targeted therapy versus sequential immunotherapy/targeted therapy as well as which regimen should be implemented as initial therapy.
Collapse
Affiliation(s)
- Anna C Pavlick
- 1 New York University Perlmutter Cancer Center, New York, NY
| | - Leslie Fecher
- 2 University of Michigan Rogel Cancer Center, Ann Arbor, MI
| | - Paolo A Ascierto
- 3 From the Instituto Nazionale Tumori, Instituto di Ricovero e Cura a Carrattere Scientifico, Fondazione G. Pascale, Naples, Italy
| | | |
Collapse
|
132
|
du Rusquec P, de Calbiac O, Robert M, Campone M, Frenel JS. Clinical utility of pembrolizumab in the management of advanced solid tumors: an evidence-based review on the emerging new data. Cancer Manag Res 2019; 11:4297-4312. [PMID: 31190995 PMCID: PMC6527794 DOI: 10.2147/cmar.s151023] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/15/2019] [Indexed: 12/11/2022] Open
Abstract
Pembrolizumab is a full-length human immunoglobulin G4 (IgG4) monoclonal antibody directed against the immune checkpoint PD-1 to remove its binding with PD-L1 and thus to restore an anti-tumor immune response of T cells. Pembrolizumab is one of the most advanced immune checkpoint inhibitors for cancer care. Apart from rare and serious adverse effects, its favorable tolerance profile enables to treat fragile patients who have often no other choice than best supportive care. The effective retained dose of pembrolizumab is a venous administration of 200 mg every 3 weeks until disease progression, intolerance or up to 24 months. Pembrolizumab has already proven its efficacy and thus obtained marketing authorization in so-called hot or hypermutated tumors or tumors expressing PD-L1 such as melanomas, non-small cell lung cancers, urothelial carcinomas, cervical cancer, etc. Pembrolizumab is also authorized in the United States in the treatment of mismatch repair-deficient tumors or with microsatellite instability. The current challenge is to expand its use in tumor types that are supposed to be less immunogenic, for example, by attempting to warm up the tumor microenvironment, or by combining pembrolizumab with other molecules. An acceptable toxicity profile of such combinations remains to explore. We review here the current indications of this drug, the main prognostic and predictive factors of its efficacy as well as the potential forthcoming indications.
Collapse
Affiliation(s)
- Pauline du Rusquec
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, Saint-Herblain44800, France
| | - Ombline de Calbiac
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, Saint-Herblain44800, France
| | - Marie Robert
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, Saint-Herblain44800, France
| | - Mario Campone
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, Saint-Herblain44800, France
| | - Jean Sebastien Frenel
- Medical Oncology Department, Institut de Cancérologie de l’Ouest, Saint-Herblain44800, France
| |
Collapse
|
133
|
Incorvaia L, Badalamenti G, Rinaldi G, Iovanna JL, Olive D, Swayden M, Terruso L, Vincenzi B, Fulfaro F, Bazan V, Russo A, Fanale D. Can the plasma PD-1 levels predict the presence and efficiency of tumor-infiltrating lymphocytes in patients with metastatic melanoma? Ther Adv Med Oncol 2019; 11:1758835919848872. [PMID: 31205506 PMCID: PMC6535916 DOI: 10.1177/1758835919848872] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The immune response in melanoma patients is locally affected by presence of tumor-infiltrating lymphocytes (TILs), generally divided into brisk, nonbrisk, and absent. Several studies have shown that a greater presence of TILs, especially brisk, in primary melanoma is associated with a better prognosis and higher survival rate. PATIENTS AND METHODS We investigated by enzyme-linked immunosorbent assay (ELISA) the correlation between PD-1 levels in plasma and the presence/absence of TILs in 28 patients with metastatic melanoma. RESULTS Low plasma PD-1 levels were correlated with brisk TILs in primary melanoma, whereas intermediate values correlated with the nonbrisk TILs, and high PD-1 levels with absent TILs. Although the low number of samples did not allow us to obtain a statistically significant correlation between the plasma PD-1 levels and the patients' overall survival depending on the absence/presence of TILs, the median survival of patients having brisk type TILs was 5 months higher than that of patients with absent and nonbrisk TILs. CONCLUSIONS This work highlights the ability of measuring the plasma PD-1 levels in order to predict the prognosis of patients with untreated metastatic melanoma without a BRAF mutation at the time of diagnosis.
Collapse
Affiliation(s)
- Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Gaetana Rinaldi
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Juan Lucio Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Mirna Swayden
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Lidia Terruso
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Bruno Vincenzi
- Medical Oncology Department, University Campus Bio-Medico, Rome, Italy
| | - Fabio Fulfaro
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
134
|
Taylor A, Rudd CE. Commentary: Small Molecule Inhibition of PD-1 Transcription is an Effective Alternative to Antibody Blockade in Cancer Therapy. ACTA ACUST UNITED AC 2019; 3:9-12. [PMID: 31111120 PMCID: PMC6525092 DOI: 10.29245/2578-3009/2019/1.1167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Alison Taylor
- Leeds Institute of Medical Research, University of Leeds, School of Medicine, Wellcome Trust Brenner Building, St James's University Hospital, LEEDS LS9 7TF, UK
| | - Christopher E Rudd
- Division of Immunology-Oncology Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada.,Département de Medicine, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
135
|
Pauken KE, Dougan M, Rose NR, Lichtman AH, Sharpe AH. Adverse Events Following Cancer Immunotherapy: Obstacles and Opportunities. Trends Immunol 2019; 40:511-523. [PMID: 31053497 DOI: 10.1016/j.it.2019.04.002] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022]
Abstract
Oncology has recently undergone a revolutionary change with widespread adoption of immunotherapy for many cancers. Immunotherapy using monoclonal antibodies against checkpoint molecules, including programmed death (PD)-1, PD ligand (PD-L)1, and cytotoxic T lymphocyte-associated antigen (CTLA)-4, is effective in a significant subset of patients. However, immune-related adverse events (irAEs) have emerged as frequent complications of checkpoint blockade, likely due to the physiological role of checkpoint pathways in regulating adaptive immunity and preventing autoimmunity. As immunotherapy becomes more common, a better understanding of the etiology of irAEs and ways to limit these events is needed. At the same time, studying these new therapy-related disorders provides an opportunity to better understand naturally occurring human autoimmune and inflammatory disorders, with the potential to improve therapies for cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Kristen E Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Noel R Rose
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
136
|
Long-Term Survival, Quality of Life, and Psychosocial Outcomes in Advanced Melanoma Patients Treated with Immune Checkpoint Inhibitors. JOURNAL OF ONCOLOGY 2019; 2019:5269062. [PMID: 31182961 PMCID: PMC6512024 DOI: 10.1155/2019/5269062] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 03/25/2019] [Indexed: 12/17/2022]
Abstract
Immune checkpoint inhibitors have become a standard of care option for the treatment of patients with advanced melanoma. Since the approval of the first immune checkpoint (CTLA-4) inhibitor ipilimumab in 2011 and programmed death-1 (PD-1) blocking monoclonal antibodies pembrolizumab and nivolumab thereafter, an increasing proportion of patients with unresectable advanced melanoma achieved long-term overall survival. Little is known about the psychosocial wellbeing, neurocognitive function, and quality of life (QOL) of these survivors. Knowledge about the long term side-effects of these novel treatments is scarce as long-term survivorship is a novel issue in the field of immunotherapy. The purpose of this review is to summarize our current knowledge regarding the survival and safety results of pivotal clinical trials in the field of advanced melanoma and to highlight potential long-term consequences that are likely to impact psychosocial wellbeing, neurocognitive functioning, and QOL. The issues raised substantiate the need for clinical investigation of these issues with the aim of optimizing comprehensive health care for advanced melanoma survivors.
Collapse
|
137
|
Coit DG, Thompson JA, Albertini MR, Barker C, Carson WE, Contreras C, Daniels GA, DiMaio D, Fields RC, Fleming MD, Freeman M, Galan A, Gastman B, Guild V, Johnson D, Joseph RW, Lange JR, Nath S, Olszanski AJ, Ott P, Gupta AP, Ross MI, Salama AK, Skitzki J, Sosman J, Swetter SM, Tanabe KK, Wuthrick E, McMillian NR, Engh AM. Cutaneous Melanoma, Version 2.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 17:367-402. [PMID: 30959471 DOI: 10.6004/jnccn.2019.0018] [Citation(s) in RCA: 284] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Cutaneous melanoma have been significantly revised over the past few years in response to emerging data on immune checkpoint inhibitor therapies and BRAF-targeted therapy. This article summarizes the data and rationale supporting extensive changes to the recommendations for systemic therapy as adjuvant treatment of resected disease and as treatment of unresectable or distant metastatic disease.
Collapse
Affiliation(s)
| | - John A Thompson
- 2Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | | | - William E Carson
- 4The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute
| | - Carlo Contreras
- 5University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | - Ryan C Fields
- 8Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine
| | - Martin D Fleming
- 9St. Jude Children's Research Hospital/The University of Tennessee Health Science Center
| | | | | | - Brian Gastman
- 12Case Comprehensive Cancer Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | - Julie R Lange
- 16The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Patrick Ott
- 19Dana-Farber/Brigham and Women's Cancer Center
| | | | | | | | | | - Jeffrey Sosman
- 20Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | | | | | | | | |
Collapse
|
138
|
Abstract
The development of new treatment options has dramatically improved the landscape for patients with advanced melanoma. Part of these advances emerged through the identification of the importance of factors that regulate the immune system, including proteins that negatively modulate T cell-mediated responses termed "immune checkpoints." Indeed, blockade of the cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) immune checkpoint served as a proof of principle that the manipulation of these molecules could induce robust anticancer effects, yet limited to a small percentage of patients. Targeting a distinct checkpoint, the PD-1 yielded improved outcomes and reduced toxicity compared with CTLA-4 blockade and, in separate studies, chemotherapy. More recently, combined CTLA-4/PD-1 blockade was shown to result in higher response rates, while accompanied by increased toxicity. In this article, we review the clinical development of anti-PD-1 monotherapy and combinations that may expand the benefit of immunotherapy for patients with advanced melanoma.
Collapse
|
139
|
Khair DO, Bax HJ, Mele S, Crescioli S, Pellizzari G, Khiabany A, Nakamura M, Harris RJ, French E, Hoffmann RM, Williams IP, Cheung A, Thair B, Beales CT, Touizer E, Signell AW, Tasnova NL, Spicer JF, Josephs DH, Geh JL, MacKenzie Ross A, Healy C, Papa S, Lacy KE, Karagiannis SN. Combining Immune Checkpoint Inhibitors: Established and Emerging Targets and Strategies to Improve Outcomes in Melanoma. Front Immunol 2019; 10:453. [PMID: 30941125 PMCID: PMC6435047 DOI: 10.3389/fimmu.2019.00453] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 02/20/2019] [Indexed: 12/13/2022] Open
Abstract
The immune system employs several checkpoint pathways to regulate responses, maintain homeostasis and prevent self-reactivity and autoimmunity. Tumor cells can hijack these protective mechanisms to enable immune escape, cancer survival and proliferation. Blocking antibodies, designed to interfere with checkpoint molecules CTLA-4 and PD-1/PD-L1 and counteract these immune suppressive mechanisms, have shown significant success in promoting immune responses against cancer and can result in tumor regression in many patients. While inhibitors to CTLA-4 and the PD-1/PD-L1 axis are well-established for the clinical management of melanoma, many patients do not respond or develop resistance to these interventions. Concerted efforts have focused on combinations of approved therapies aiming to further augment positive outcomes and survival. While CTLA-4 and PD-1 are the most-extensively researched targets, results from pre-clinical studies and clinical trials indicate that novel agents, specific for checkpoints such as A2AR, LAG-3, IDO and others, may further contribute to the improvement of patient outcomes, most likely in combinations with anti-CTLA-4 or anti-PD-1 blockade. This review discusses the rationale for, and results to date of, the development of inhibitory immune checkpoint blockade combination therapies in melanoma. The clinical potential of new pipeline therapeutics, and possible future therapy design and directions that hold promise to significantly improve clinical prognosis compared with monotherapy, are discussed.
Collapse
Affiliation(s)
- Duaa O. Khair
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Heather J. Bax
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer & Pharmaceutical Sciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Silvia Mele
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Silvia Crescioli
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Giulia Pellizzari
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Atousa Khiabany
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Mano Nakamura
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | | | - Elise French
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Ricarda M. Hoffmann
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer & Pharmaceutical Sciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Iwan P. Williams
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Anthony Cheung
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, Guy's Cancer Centre, King's College London, London, United Kingdom
| | - Benjamin Thair
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Charlie T. Beales
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Emma Touizer
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Adrian W. Signell
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Nahrin L. Tasnova
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - James F. Spicer
- School of Cancer & Pharmaceutical Sciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Debra H. Josephs
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
- School of Cancer & Pharmaceutical Sciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Jenny L. Geh
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Alastair MacKenzie Ross
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Ciaran Healy
- Department of Plastic Surgery at Guy's, King's, and St. Thomas' Hospitals, London, United Kingdom
| | - Sophie Papa
- School of Cancer & Pharmaceutical Sciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Katie E. Lacy
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| | - Sophia N. Karagiannis
- St. John's Institute of Dermatology, School of Basic & Medical Biosciences, Guy's Hospital, King's College London, London, United Kingdom
| |
Collapse
|
140
|
Rotte A, Jin JY, Lemaire V. Mechanistic overview of immune checkpoints to support the rational design of their combinations in cancer immunotherapy. Ann Oncol 2019; 29:71-83. [PMID: 29069302 DOI: 10.1093/annonc/mdx686] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Checkpoint receptor blockers, known to act by blocking the pathways that inhibit immune cell activation and stimulate immune responses against tumor cells, have been immensely successful in the treatment of cancer. Among several checkpoint receptors of immune cells, cytotoxic T-lymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), T-cell immunoglobulin and ITIM domain (TIGIT), T-cell immunoglobulin-3 (TIM-3) and lymphocyte activation gene 3 (LAG-3) are the most commonly targeted checkpoints for cancer immunotherapy. Six drugs including one CTLA-4 blocker (ipilimumab), two PD-1 blockers (nivolumab and pembrolizumab) and three PD-L1 blockers (atezolizumab, avelumab and durvalumab) are approved for the treatment of different types of cancers including both solid tumors such as melanoma, lung cancer, head and neck cancer, bladder cancer and Merkel cell cancer as well as hematological tumors such as classic Hodgkin's lymphoma. The main problem with checkpoint blockers is that only a fraction of patients respond to the therapy. Insufficient immune activation is considered as one of the main reason for low response rates and combination of checkpoint blockers has been proposed to increase the response rates. The combination of checkpoint blockers was successful in melanoma but had significant adverse events. A combination that is selected based on the mechanistic differences between checkpoints and the differences in expression of checkpoints and their ligands in the tumor microenvironment could have a synergistic effect in a given cancer subtype and also have a manageable safety profile. This review aims to help in design of optimal checkpoint blocker combinations by discussing the mechanistic details and outlining the subtle differences between major checkpoints targeted for cancer immunotherapy.
Collapse
Affiliation(s)
- A Rotte
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, USA
| | - J Y Jin
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, USA
| | - V Lemaire
- Department of Clinical Pharmacology, Genentech Research and Early Development, South San Francisco, USA
| |
Collapse
|
141
|
Thompson JA, Schneider BJ, Brahmer J, Andrews S, Armand P, Bhatia S, Budde LE, Costa L, Davies M, Dunnington D, Ernstoff MS, Frigault M, Hoffner B, Hoimes CJ, Lacouture M, Locke F, Lunning M, Mohindra NA, Naidoo J, Olszanski AJ, Oluwole O, Patel SP, Reddy S, Ryder M, Santomasso B, Shofer S, Sosman JA, Wahidi M, Wang Y, Johnson-Chilla A, Scavone JL. Management of Immunotherapy-Related Toxicities, Version 1.2019, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2019; 17:255-289. [DOI: 10.6004/jnccn.2019.0013] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aim of the NCCN Guidelines for Management of Immunotherapy-Related Toxicities is to provide guidance on the management of immune-related adverse events resulting from cancer immunotherapy. The NCCN Management of Immunotherapy-Related Toxicities Panel is an interdisciplinary group of representatives from NCCN Member Institutions and ASCO, consisting of medical and hematologic oncologists with expertise in a wide array of disease sites, and experts from the fields of dermatology, gastroenterology, neuro-oncology, nephrology, emergency medicine, cardiology, oncology nursing, and patient advocacy. Several panel representatives are members of the Society for Immunotherapy of Cancer (SITC). The initial version of the NCCN Guidelines was designed in general alignment with recommendations published by ASCO and SITC. The content featured in this issue is an excerpt of the recommendations for managing toxicity related to immune checkpoint blockade and a review of existing evidence. For the full version of the NCCN Guidelines, including recommendations for managing toxicities related to chimeric antigen receptor T-cell therapy, visitNCCN.org.
Collapse
Affiliation(s)
- John A. Thompson
- 1Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Julie Brahmer
- 3The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | - Shailender Bhatia
- 1Fred Hutchinson Cancer Research Center/Seattle Cancer Care Alliance
| | | | - Luciano Costa
- 7University of Alabama at Birmingham Comprehensive Cancer Center
| | | | | | | | | | | | - Christopher J. Hoimes
- 13Case Comprehensive Center/University Hospitals Seidman Cancer Center and Cleveland Clinic Taussig Cancer Institute
| | | | | | | | - Nisha A. Mohindra
- 16Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | - Jarushka Naidoo
- 3The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | | | | | | | | | | | | | - Jeffrey A. Sosman
- 16Robert H. Lurie Comprehensive Cancer Center of Northwestern University
| | | | - Yinghong Wang
- 23The University of Texas MD Anderson Cancer Center; and
| | | | | |
Collapse
|
142
|
Zhang B, Zhou YL, Chen X, Wang Z, Wang Q, Ju F, Ren S, Xu R, Xue Q, Wu Q. Efficacy and safety of CTLA-4 inhibitors combined with PD-1 inhibitors or chemotherapy in patients with advanced melanoma. Int Immunopharmacol 2019; 68:131-136. [DOI: 10.1016/j.intimp.2018.12.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 11/26/2022]
|
143
|
Heinzerling L, de Toni EN, Schett G, Hundorfean G, Zimmer L. Checkpoint Inhibitors. DEUTSCHES ARZTEBLATT INTERNATIONAL 2019; 116:119-126. [PMID: 30940340 PMCID: PMC6454802 DOI: 10.3238/arztebl.2019.0119] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 05/05/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Treatment with checkpoint inhibitors such as anti-programmed death-1 (anti-PD-1), anti-PD-ligand 1 (anti-PD-L1), and anti-cytotoxic T-lymphocyte antigen-4 (anti-CTLA-4) antibodies can prolong the survival of cancer patients, but it also induces autoimmune side effects in 86-96% of patients by activating the immune system. In 17-59% of patients, these are severe or even life-threatening. METHODS This review is based on pertinent articles retrieved by a search in PubMed and on an evaluation of a side-effect registry. RESULTS Checkpoint-inhibitor-induced autoimmune side effects manifest themselves in all organ systems, most commonly as skin lesions (46-62%), autoimmune colitis (22-48%), autoimmune hepatitis (7-33%), and endocrinopathies (thyroiditis, hypophysitis, adrenalitis, diabetes mellitus; 12-34%). Rarer side effects include pneumonitis (3-8%), nephritis (1-7%), cardiac side effects including cardiomyositis (5%), and neurological side effects (1-5%). Severe (sometimes lethal) side effects arise in 17-21%, 20-28%, and 59% of patients undergoing anti-PD-1 and anti- CTLA-4 antibody treatment and the approved combination therapy, respectively. With proper monitoring, however, these side effects can be recognized early and, usually, treated with success. Endocrine side effects generally require long-term hormone substitution. Patients who have stopped taking checkpoint inhibitors because of side effects do not show a poorer response of their melanoma or shorter survival in comparison to patients who continue to take checkpoint inhibitors. CONCLUSION The complex management of checkpoint-inhibitor-induced side effects should be coordinated in experienced centers. The creation of an interdisciplinary "tox team" with designated experts for organ-specific side effects has proven useful. Prospective registry studies based on structured documentation of side effects in routine clinical practice are currently lacking and urgently needed.
Collapse
Affiliation(s)
| | - Enrico N. de Toni
- Department of Internal Medicine II, University Hospital, Ludwig-Maximilians-University (LMU) Munich
| | - Georg Schett
- Department of Medicine 3, University Hospital Erlangen-Nürnberg
| | | | - Lisa Zimmer
- Clinic for Dermatology, Essen University Hospital, University of Duisburg-Essen
| |
Collapse
|
144
|
Asher N, Marom EM, Ben-Betzalel G, Baruch EN, Steinberg-Silman Y, Schachter J, Shapira-Frommer R, Markel G. Recurrent Pneumonitis in Patients with Melanoma Treated with Immune Checkpoint Inhibitors. Oncologist 2019; 24:640-647. [PMID: 30777894 DOI: 10.1634/theoncologist.2018-0352] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 01/11/2019] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have changed the oncologic landscape in the past few years. Alongside impressive antitumor responses, new novel immune-related adverse events (irAEs) have emerged; pneumonitis is an irAE that can potentially be fatal and necessitates a proper management. No consensus exists regarding steroid treatment duration or drug rechallenge options. Our study describes the clinical and radiological course of melanoma patients diagnosed with immune-related pneumonitis that has recurred because of rechallenge attempt or despite complete treatment discontinuation (unprovoked). MATERIALS AND METHODS The study population was composed of patients with metastatic melanoma who were treated with anti-programmed cell death 1 (PD-1) as monotherapy or in combination with anti-cytotoxic T lymphocyte antigen-4 and who were diagnosed with immune-related pneumonitis. For recurrent cases after clinical and radiological resolution, we explored the differences from cases with no recurrence. RESULTS Nineteen out of 386 (4.8%) patients treated with ICI were diagnosed with pneumonitis. Median age was 66 years, and 53% were male. Compared with single-agent nivolumab, patients treated with ipilimumab-nivolumab combination presented with an earlier onset (27.5 vs. 10.3 weeks, respectively, p = .015) and had higher grades of severity. After complete resolution, rechallenge was attempted in seven patients; three of them had recurrent pneumonitis. Three other patients experienced recurrent pneumonitis despite complete discontinuation of the drug (unprovoked by rechallenge). The latter were characterized with an earlier onset of the first pneumonitis compared with those who did not experience recurrence (median, 12.4 vs. 26.4 weeks) and a shorter course of steroid treatment at first episode (median, 5.1 vs. 10 weeks). Recurrent cases were generally more severe than the first episode. CONCLUSION Unprovoked recurrent pneumonitis is a new, poorly reported entity that requires further investigation. Our observations suggest that cases of pneumonitis that present early in the course of immunotherapy treatment may recur despite treatment discontinuation, thus necessitating closer monitoring and a longer course of steroid treatment. IMPLICATIONS FOR PRACTICE This article sheds light on a poorly described immune-related adverse event: recurrent pneumonitis despite complete discontinuation of immunotherapy (unprovoked), in patients with advanced melanoma.
Collapse
Affiliation(s)
- Nethanel Asher
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Edith M Marom
- Institute for Radiology, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Ben-Betzalel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Erez Nissim Baruch
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Steinberg-Silman
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
| | - Jacob Schachter
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Gal Markel
- Ella Lemelbaum Institute for Immuno-Oncology, Sheba Medical Center, Ramat Gan, Israel
- Talpiot Medical Leadership Program, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
145
|
Recent success and limitations of immune checkpoint inhibitors for cancer: a lesson from melanoma. Virchows Arch 2019; 474:421-432. [PMID: 30747264 DOI: 10.1007/s00428-019-02538-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 01/20/2019] [Accepted: 02/01/2019] [Indexed: 02/08/2023]
Abstract
Several researches have been carried over the last few decades to understand of how cancer evades the immune system and thus to identify therapies that could directly act on patient's immune system in the way of restore or induce a response to cancer. As a consequence, "cancer immunotherapy" is conquering predominantly the modern scenario of the fight against cancer. The recent clinical success of immune checkpoint inhibitors (ICIs) has created an entire new class of anti-cancer drugs and restored interest in the field of immuno-oncology, leading to regulatory approvals of several agents for the treatment of a variety of malignancies. The first to be approved in 2011 was the anti-CTLA-4 antibody ipilimumab for the treatment of unresectable or metastatic melanoma. Subsequently, the anti-PD-1s, nivolumab and pembrolizumab, received regulatory approvals for the treatment of melanoma and several other cancers. More recently, three anti-PD-L1 antibodies have received approval: atezolizumab and durvalumab for locally advanced or metastatic urothelial carcinoma and metastatic non-small cell lung cancer (NSCLC) and avelumab for the treatment of locally advanced or metastatic urothelial carcinoma and metastatic Merkel cell carcinoma. This review, starting from the results of melanoma trials, highlights in turn different ICIs and data for different indications in several malignancies are included under each drug class.
Collapse
|
146
|
Castinetti F, Albarel F, Archambeaud F, Bertherat J, Bouillet B, Buffier P, Briet C, Cariou B, Caron P, Chabre O, Chanson P, Cortet C, Do Cao C, Drui D, Haissaguerre M, Hescot S, Illouz F, Kuhn E, Lahlou N, Merlen E, Raverot V, Smati S, Verges B, Borson-Chazot F. French Endocrine Society Guidance on endocrine side effects of immunotherapy. Endocr Relat Cancer 2019; 26:G1-G18. [PMID: 30400055 PMCID: PMC6347286 DOI: 10.1530/erc-18-0320] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/08/2018] [Indexed: 12/14/2022]
Abstract
The management of cancer patients has changed due to the considerably more frequent use of immune checkpoint inhibitors (ICPIs). However, the use of ICPI has a risk of side effects, particularly endocrine toxicity. Since the indications for ICPI are constantly expanding due to their efficacy, it is important that endocrinologists and oncologists know how to look for this type of toxicity and how to treat it when it arises. In view of this, the French Endocrine Society initiated the formulation of a consensus document on ICPI-related endocrine toxicity. In this paper, we will introduce data on the general pathophysiology of endocrine toxicity, and we will then outline expert opinion focusing primarily on methods for screening, management and monitoring for endocrine side effects in patients treated by ICPI. We will then look in turn at endocrinopathies that are induced by ICPI including dysthyroidism, hypophysitis, primary adrenal insufficiency and fulminant diabetes. In each chapter, expert opinion will be given on the diagnosis, management and monitoring for each complication. These expert opinions will also discuss the methodology for categorizing these side effects in oncology using 'common terminology criteria for adverse events' (CTCAE) and the difficulties in applying this to endocrine side effects in the case of these anti-cancer therapies. This is shown in particular by certain recommendations that are used for other side effects (high-dose corticosteroids, contraindicated in ICPI for example) and that cannot be considered as appropriate in the management of endocrine toxicity, as it usually does not require ICPI withdrawal or high-dose glucocorticoid intake.
Collapse
Affiliation(s)
- F Castinetti
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), and Department of Endocrinology, Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital de la Conception, Centre de Référence des Maladies Rares de l’Hypophyse HYPO, Marseille, France
- Correspondence should be addressed to F Castinetti:
| | - F Albarel
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale (INSERM), U1251, Marseille Medical Genetics (MMG), and Department of Endocrinology, Assistance Publique-Hôpitaux de Marseille (AP-HM), Hôpital de la Conception, Centre de Référence des Maladies Rares de l’Hypophyse HYPO, Marseille, France
| | - F Archambeaud
- Service de Médecine Interne B – Endocrinologie, Limoges Cedex, France
| | - J Bertherat
- Hôpital Cochin, Service d’Endocrinologie et Maladies Métaboliques, Paris Cedex 14, France
| | - B Bouillet
- CHU Dijon, Hôpital François Mitterrand, Service d’Endocrinologie, Diabétologie, Maladies Métaboliques, Dijon Cedex, France
- Unité INSERM LNC-UMR 1231, Université de Bourgogne, Dijon, France
| | - P Buffier
- CHU Dijon, Hôpital François Mitterrand, Service d’Endocrinologie, Diabétologie, Maladies Métaboliques, Dijon Cedex, France
| | - C Briet
- Institut MITOVASC, INSERM U1083, Angers University, Department of Endocrinology, Diabetology and Nutrition, University Medical Center, Angers, France
| | - B Cariou
- Department of Endocrinology, L’Institut du Thorax, CHU Nantes, Nantes, France
| | - Ph Caron
- CHU de Toulouse – Hôpital Larrey – Service d’Endocrinologie – Maladies métaboliques – Nutrition, TSA 30030, Toulouse Cedex 9, France
| | - O Chabre
- CHU de Grenoble – Hôpital Albert Michallon, Service d’Endocrinologie-Diabétologie-Nutrition, Grenoble Cedex 9, France
| | - Ph Chanson
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital de Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, et UMR S-1185 Faculté de Médecine Paris-Sud, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - C Cortet
- CHRU de Lille – Hopital Huriez, Service d’Endocrinologie, Lille Cedex, France
| | - C Do Cao
- CHRU de Lille – Hopital Huriez, Service d’Endocrinologie, Lille Cedex, France
| | - D Drui
- Department of Endocrinology, L’Institut du Thorax, CHU Nantes, Nantes, France
| | - M Haissaguerre
- CHU de Bordeaux – Hôpital du Haut Lévêque, Service d’Endocrinologie-Diabétologie et Maladies Métaboliques, Pessac Cedex, France
| | - S Hescot
- Institut Curie, Oncologie Endocrinienne, Saint Cloud, France
| | - F Illouz
- Department of Endocrinology, Diabetes and Nutrition, Reference Centre of Rare Thyroid Disease, Hospital of Angers, Angers Cedex 09, France
| | - E Kuhn
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital de Bicêtre, Service d’Endocrinologie et des Maladies de la Reproduction, et UMR S-1185 Faculté de Médecine Paris-Sud, University of Paris-Saclay, Le Kremlin-Bicêtre, France
| | - N Lahlou
- Département d’Hormonologie Spécialisée, BPR-AS, Pannes, France
| | - E Merlen
- CHRU de Lille – Hopital Huriez, Service d’Endocrinologie, Lille Cedex, France
| | - V Raverot
- Hospices Civils de Lyon, Laboratoire d’Hormonologie, Service de Biochimie et Biologie Moléculaire, Groupement Hospitalier Est, Lyon, France
| | - S Smati
- Department of Endocrinology, L’Institut du Thorax, CHU Nantes, Nantes, France
| | - B Verges
- CHU Dijon, Hôpital François Mitterrand, Service d’Endocrinologie, Diabétologie, Maladies Métaboliques, Dijon Cedex, France
- Unité INSERM LNC-UMR 1231, Université de Bourgogne, Dijon, France
| | - F Borson-Chazot
- Hospices Civils de Lyon, Fédération d’Endocrinologie, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
147
|
Carreau NA, Pavlick AC. Nivolumab and ipilimumab: immunotherapy for treatment of malignant melanoma. Future Oncol 2019; 15:349-358. [DOI: 10.2217/fon-2018-0607] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
As recently as 10 years ago, a diagnosis of metastatic melanoma was considered fatal, with a prognosis of typically 6 months or less from diagnosis. The development of checkpoint inhibitors, such as ipilimumab and nivolumab, which modulate the effects of the CTLA-4 and PD-1, respectively, has revolutionized outcomes for these patients. Monotherapy improves metastatic disease survival, but dual therapy provides greater benefit with 58% of patients alive at 3 years. Combination immunotherapy is even active in brain metastases. In the adjuvant setting, data show that at 1 year over 70% patients remain disease-free with PD-1 blockade. Immunotherapy is generally safe and well tolerated. However, treatment-related endocrinopathies require long-term medications. Nowadays, advanced cutaneous melanoma is a more manageable disease.
Collapse
Affiliation(s)
- Nicole A Carreau
- Department of Medical Oncology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Anna C Pavlick
- Department of Medical Oncology, New York University Langone Medical Center, New York, NY 10016, USA
| |
Collapse
|
148
|
Conroy JM, Pabla S, Nesline MK, Glenn ST, Papanicolau-Sengos A, Burgher B, Andreas J, Giamo V, Wang Y, Lenzo FL, Bshara W, Khalil M, Dy GK, Madden KG, Shirai K, Dragnev K, Tafe LJ, Zhu J, Labriola M, Marin D, McCall SJ, Clarke J, George DJ, Zhang T, Zibelman M, Ghatalia P, Araujo-Fernandez I, de la Cruz-Merino L, Singavi A, George B, MacKinnon AC, Thompson J, Singh R, Jacob R, Kasuganti D, Shah N, Day R, Galluzzi L, Gardner M, Morrison C. Next generation sequencing of PD-L1 for predicting response to immune checkpoint inhibitors. J Immunother Cancer 2019; 7:18. [PMID: 30678715 PMCID: PMC6346512 DOI: 10.1186/s40425-018-0489-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND PD-L1 immunohistochemistry (IHC) has been traditionally used for predicting clinical responses to immune checkpoint inhibitors (ICIs). However, there are at least 4 different assays and antibodies used for PD-L1 IHC, each developed with a different ICI. We set to test if next generation RNA sequencing (RNA-seq) is a robust method to determine PD-L1 mRNA expression levels and furthermore, efficacy of predicting response to ICIs as compared to routinely used, standardized IHC procedures. METHODS A total of 209 cancer patients treated on-label by FDA-approved ICIs, with evaluable responses were assessed for PD-L1 expression by RNA-seq and IHC, based on tumor proportion score (TPS) and immune cell staining (ICS). A subset of serially diluted cases was evaluated for RNA-seq assay performance across a broad range of PD-L1 expression levels. RESULTS Assessment of PD-L1 mRNA levels by RNA-seq demonstrated robust linearity across high and low expression ranges. PD-L1 mRNA levels assessed by RNA-seq and IHC (TPS and ICS) were highly correlated (p < 2e-16). Sub-analyses showed sustained correlation when IHC results were classified as high or low by clinically accepted cut-offs (p < 0.01), and results did not differ by tumor type or anti-PD-L1 antibody used. Overall, a combined positive PD-L1 result (≥1% IHC TPS and high PD-L1 expression by RNA-Seq) was associated with a 2-to-5-fold higher overall response rate (ORR) compared to a double negative result. Standard assessments of sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) showed that a PD-L1 positive assessment for melanoma samples by RNA-seq had the lowest sensitivity (25%) but the highest PPV (72.7%). Among the three tumor types analyzed in this study, the only non-overlapping confidence interval for predicting response was for "RNA-seq low vs high" in melanoma. CONCLUSIONS Measurement of PD-L1 mRNA expression by RNA-seq is comparable to PD-L1 expression by IHC both analytically and clinically in predicting ICI response. RNA-seq has the added advantages of being amenable to standardization and avoidance of interpretation bias. PD-L1 by RNA-seq needs to be validated in future prospective ICI clinical studies across multiple histologies.
Collapse
Affiliation(s)
- Jeffrey M Conroy
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Sarabjot Pabla
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Mary K Nesline
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Sean T Glenn
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | | | - Blake Burgher
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
| | | | - Vincent Giamo
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Yirong Wang
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
| | | | - Wiam Bshara
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Maya Khalil
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Grace K Dy
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | | | - Keisuke Shirai
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | | | - Laura J Tafe
- Dartmouth-Hitchcock Medical Center, Lebanon, NH, 03756, USA
| | - Jason Zhu
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Matthew Labriola
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Daniele Marin
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Shannon J McCall
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Jeffrey Clarke
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Daniel J George
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Tian Zhang
- Duke University Medical Center, 905 S. Lasalle Street, Durham, NC, 27710, USA
| | - Matthew Zibelman
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA
| | - Pooja Ghatalia
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA
| | | | | | - Arun Singavi
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Ben George
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | | | - Jonathan Thompson
- Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI, 53226, USA
| | - Rajbir Singh
- Meharry Medical College, 1005 Dr DB Todd Jr Blvd, Nashville, TN, 37208, USA
| | - Robin Jacob
- Meharry Medical College, 1005 Dr DB Todd Jr Blvd, Nashville, TN, 37208, USA
| | | | - Neel Shah
- Community Hospital, Munster, IN, 46321, USA
| | - Roger Day
- University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, 10065, USA
- Université Paris Descartes/Paris V, 75006, Paris, France
| | - Mark Gardner
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA
| | - Carl Morrison
- OmniSeq, Inc., 700 Ellicott Street, Buffalo, NY, 14203, USA.
- Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
149
|
Immunotherapy: enhancing the efficacy of this promising therapeutic in multiple cancers. Clin Sci (Lond) 2019; 133:181-193. [PMID: 30659159 DOI: 10.1042/cs20181003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 12/31/2022]
Abstract
Cancer treatments often reach a refractory period leading to treatment failure and patients developing disease recurrence. This can be due to tumour cells escaping the immune response and creating an immunosuppressive microenvironment enhancing cancer progression. Immunotherapy has become a promising tool for cancer treatment as it restores the anti-tumour response of the patient's immune system. Immune checkpoint inhibitors are the most widely studied immunotherapies worldwide and are now approved for multiple cancers. However, chimeric antigen receptor (CAR)-T cell therapy has also shown promise by targeting T lymphocytes that are genetically modified ex vivo to express CARs and this is now approved to treat some haematological cancers. Although immunotherapy has shown successful treatment outcomes in multiple cancers, some patients do not respond to this treatment. Therefore, approaches to enhance the efficacy of immunotherapies are likely to be the key to improve their effectiveness. Therefore, combination therapies of checkpoint inhibitors +/- chemotherapy are at the forefront of current research. Furthermore, biomarkers that predict treatment response are now beginning to emerge. Additionally, utilising nanoparticles as a newly targeted drug delivery system to enhance CAR-T cell therapy may enhance the efficacy of the cells when re-infused within the patient. Even if efficacy is enhanced, severe immune-related adverse events (irAEs) occur that are life-threatening and could lead to therapy being stopped. Therefore, predictive biomarkers for toxicity are also needed to improve both the patient's quality of life and treatment outcomes. This review will look at the current immunotherapies in clinical trials and discuss how to enhance their efficacy.
Collapse
|
150
|
Tarhini AA, Lee SJ, Li X, Rao UNM, Nagarajan A, Albertini MR, Mitchell JW, Wong SJ, Taylor MA, Laudi N, Truong PV, Conry RM, Kirkwood JM. E3611-A Randomized Phase II Study of Ipilimumab at 3 or 10 mg/kg Alone or in Combination with High-Dose Interferon-α2b in Advanced Melanoma. Clin Cancer Res 2019; 25:524-532. [PMID: 30420448 PMCID: PMC6335150 DOI: 10.1158/1078-0432.ccr-18-2258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/30/2018] [Accepted: 11/02/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE Interferon-α favors a Th1 shift in immunity, and combining with ipilimumab (ipi) at 3 or 10 mg/kg may downregulate CTLA4-mediated suppressive effects, leading to more durable antitumor immune responses. A study of tremelimumab and high-dose interferon-α (HDI) showed promising efficacy, supporting this hypothesis. PATIENTS AND METHODS E3611 followed a 2-by-2 factorial design (A: ipi10+HDI; B: ipi10; C: ipi3+HDI; D: ipi3) to evaluate (i) no HDI versus HDI (across ipilimumab doses) and (ii) ipi3 versus ipi10 (across HDI status). We hypothesized that median progression-free survival (PFS) would improve from 3 to 6 months with HDI versus no HDI and with ipi10 versus ipi3. RESULTS For eligible and treated patients (N = 81) at a median follow-up time of 29.8 months, median PFS was 4.4 months [95% confidence interval (CI), 2.7-8.2] when ipilimumab was used alone and 7.5 months (95% CI, 5.1-11.0) when HDI was added. Median PFS was 3.8 months (95% CI, 2.6-7.5) with 3 mg/kg ipilimumab and 6.5 months (95% CI, 5.1-13.5) with 10 mg/kg. By study arm, median PFS was 8.0 months (95% CI, 2.8-20.2) in arm A, 6.2 months (95% CI, 2.7-25.7) in B, 5.7 months (95% CI, 1.5-11.1) in C, and 2.8 months (95% CI, 2.6-5.7) in D. The differences in PFS and overall survival (OS) did not reach statistical significance. Adverse events were consistent with the known profiles of ipilimumab and HDI and significantly higher with HDI and ipi10. CONCLUSIONS Although PFS was increased, the differences resulting from adding interferon-α or a higher dose of ipilimumab did not reach statistical significance and do not outweigh the added toxicity risks.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Cleveland Clinic and Case Comprehensive Cancer Center, Cleveland, Ohio.
- University of Pittsburgh Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Sandra J Lee
- ECOG-ACRIN Biostatistics Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Xiaoxue Li
- Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Uma N M Rao
- University of Pittsburgh Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Mark R Albertini
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | | | | | - Mark A Taylor
- Lewis Cancer and Research Pavilion at St. Joseph's, Savannah, Georgia
| | - Noel Laudi
- Minnesota Oncology Hematology PA, Minneapolis, Minnesota
| | | | - Robert M Conry
- University of Alabama at Birmingham, Birmingham, Alabama
| | - John M Kirkwood
- University of Pittsburgh Cancer Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| |
Collapse
|