101
|
Shah MA, Udrea AA, Bondarenko I, Mansoor W, Sánchez RG, Sarosiek T, Bozzarelli S, Schenker M, Gomez-Martin C, Morgan C, Özgüroğlu M, Pikiel J, Kalofonos HP, Wojcik E, Buchler T, Swinson D, Cicin I, Joseph M, Vynnychenko I, Luft AV, Enzinger PC, Salek T, Papandreou C, Tournigand C, Maiello E, Wei R, Ferry D, Gao L, Oliveira JM, Ajani JA. Evaluating Alternative Ramucirumab Doses as a Single Agent or with Paclitaxel in Second-Line Treatment of Locally Advanced or Metastatic Gastric/Gastroesophageal Junction Adenocarcinoma: Results from Two Randomized, Open-Label, Phase II Studies. Cancers (Basel) 2022; 14:cancers14051168. [PMID: 35267477 PMCID: PMC8909008 DOI: 10.3390/cancers14051168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Ramucirumab is indicated at a dosage of 8 mg/kg every 2 weeks as monotherapy or in combination with paclitaxel for second-line advanced/metastatic gastric/gastroesophageal junction (GEJ) adenocarcinoma. A post hoc analysis of the phase III trials REGARD and RAINBOW suggested a positive correlation between ramucirumab exposure and efficacy. Studies JVDB and JVCZ explored different ramucirumab dosing regimens as monotherapy and in combination with paclitaxel, respectively. Here we report results from these studies, in which JVDB evaluated the pharmacokinetics and safety of the currently registered dosing regimen for ramucirumab monotherapy and three exploratory dosing regimens, and JVCZ evaluated the efficacy and safety of a higher dosing regimen of ramucirumab in combination with paclitaxel in second-line gastric/GEJ adenocarcinoma. Overall, the safety profiles were similar between the registered dose and the exploratory dosing regimens. However, a lack of a dose/exposure-response relationship supports the standard dose of ramucirumab as second-line treatment for patients with advanced/metastatic gastric/GEJ adenocarcinoma. Abstract Studies JVDB and JVCZ examined alternative ramucirumab dosing regimens as monotherapy or combined with paclitaxel, respectively, in patients with advanced/metastatic gastric/gastroesophageal junction (GEJ) adenocarcinoma. For JVDB, randomized patients (N = 164) received ramucirumab monotherapy at four doses: 8 mg/kg every 2 weeks (Q2W) (registered dose), 12 mg/kg Q2W, 6 mg/kg weekly (QW), or 8 mg/kg on days 1 and 8 (D1D8) every 3 weeks (Q3W). The primary objectives were the safety and pharmacokinetics of ramucirumab monotherapy. For JVCZ, randomized patients (N = 245) received paclitaxel (80 mg/m2-D1D8D15) plus ramucirumab (8 mg/kg- or 12 mg/kg-Q2W). The primary objective was progression-free survival (PFS) of 12 mg/kg-Q2W arm versus placebo from RAINBOW using meta-analysis. Relative to the registered dose, exploratory dosing regimens (EDRs) led to higher ramucirumab serum concentrations in both studies. EDR safety profiles were consistent with previous studies. In JVDB, serious adverse events occurred more frequently in the 8 mg/kg-D1D8-Q3W arm versus the registered dose; 6 mg/kg-QW EDR had a higher incidence of bleeding/hemorrhage. In JVCZ, PFS was improved with the 12 mg/kg plus paclitaxel combination versus placebo in RAINBOW; however, no significant PFS improvement was observed between the 12 mg/kg and 8 mg/kg arms. The lack of a dose/exposure-response relationship in these studies supports the standard dose of ramucirumab 8 mg/kg-Q2W as monotherapy or in combination with paclitaxel as second-line treatment for advanced/metastatic gastric/GEJ adenocarcinoma.
Collapse
Affiliation(s)
- Manish A. Shah
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY 10021, USA
- Correspondence: ; Tel.: +1-646-962-6200
| | | | - Igor Bondarenko
- Department of Oncology, Dnipropetrovsk Medical Academy, 49044 Dnipropetrovsk, Ukraine;
| | - Was Mansoor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester M20 4BX, UK;
| | - Raquel Guardeño Sánchez
- Department of Medical Oncology, Catalan Institute of Oncology (ICO) Girona Hospital Dr Josep Trueta, 17007 Girona, Spain;
| | - Tomasz Sarosiek
- Department of Clinical Oncology and Oncological Surgery, LUXMED Onkologia, 04125 Warszawa, Poland;
| | - Silvia Bozzarelli
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, 20089 Milan, Italy;
| | - Michael Schenker
- Centrul de Oncologie Sf. Nectarie SRL, 200542 Craiova, Romania;
- Department of Medical Oncology, University of Medicine and Pharmacy Craiova, 200342 Craiova, Romania
| | - Carlos Gomez-Martin
- Medical Oncology Department, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain;
| | - Carys Morgan
- Department of Clinical Oncology, Velindre Cancer Centre, Cardiff CF14 2TL, UK;
| | - Mustafa Özgüroğlu
- Medical Oncology, Istanbul University, Cerrahpaşa, Fatih, Istanbul 34098, Turkey;
| | - Joanna Pikiel
- Department of Oncology, Copernicus Podmiot Leczniczy, 80-803 Gdańsk, Poland;
| | - Haralabos P. Kalofonos
- Department of Oncology, University General Hospital of Patras Rion, 26504 Patras, Greece;
| | | | - Tomas Buchler
- Department of Oncology, First Faculty of Medicine, Charles University and Thomayer University Hospital, 14059 Prague, Czech Republic;
| | - Daniel Swinson
- Institute of Oncology, St James’s University Hospital, Leeds LS9 7TF, UK;
| | - Irfan Cicin
- Medical Oncology, Trakya University, Edirne 22030, Turkey;
| | - Mano Joseph
- Deanesly Centre, New Cross Hospital, Wolverhamptom WV10 0QP, UK;
| | - Ihor Vynnychenko
- Sumy Regional Oncology Center, Sumy State University, 40000 Sumy, Ukraine;
| | - Alexander Valerievich Luft
- Department of Oncology No 1 (Thoracic Surgery), Leningrad Regional Clinical Hospital, 194291 St. Petersburg, Russia;
| | - Peter C. Enzinger
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
| | - Tomas Salek
- Department of Clinical Oncology, Narodny Onkologicky Ustav, 83310 Bratislava, Slovakia;
| | - Christos Papandreou
- Department of Medical Oncology, Faculty of Medicine, University of Thessaly, Biopolis, 41223 Larissa, Greece;
| | - Christophe Tournigand
- Department of Medical Oncology, Henri Mondor et Albert Chenevier Teaching Hospital, Assistance Publique-Hôpitaux de Paris, University of Paris-Est Creteil, 94000 Créteil, France;
| | - Evaristo Maiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, Viale Cappuccini 1, 71013 San Giovanni Rotondo, Italy;
| | - Ran Wei
- Eli Lilly and Company, Indianapolis, IN 46225, USA;
| | - David Ferry
- Eli Lilly and Company, New York, NY 10016, USA; (D.F.); (L.G.); (J.M.O.)
| | - Ling Gao
- Eli Lilly and Company, New York, NY 10016, USA; (D.F.); (L.G.); (J.M.O.)
| | - Joana M. Oliveira
- Eli Lilly and Company, New York, NY 10016, USA; (D.F.); (L.G.); (J.M.O.)
| | - Jaffer A. Ajani
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| |
Collapse
|
102
|
Lee PS, MacDonald KG, Massi E, Chew PV, Bee C, Perkins P, Chau B, Thudium K, Lohre J, Nandi P, Deyanova EG, Barman I, Gudmundsson O, Dollinger G, Sproul T, Engelhardt JJ, Strop P, Rajpal A. Improved therapeutic index of an acidic pH-selective antibody. MAbs 2022; 14:2024642. [PMID: 35192429 PMCID: PMC8865267 DOI: 10.1080/19420862.2021.2024642] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Although therapeutically efficacious, ipilimumab can exhibit dose-limiting toxicity that prevents maximal efficacious clinical outcomes and can lead to discontinuation of treatment. We hypothesized that an acidic pH-selective ipilimumab (pH Ipi), which preferentially and reversibly targets the acidic tumor microenvironment over the neutral periphery, may have a more favorable therapeutic index. While ipilimumab has pH-independent CTLA-4 affinity, pH Ipi variants have been engineered to have up to 50-fold enhanced affinity to CTLA-4 at pH 6.0 compared to pH 7.4. In hCTLA-4 knock-in mice, these variants have maintained anti-tumor activity and reduced peripheral activation, a surrogate marker for toxicity. pH-sensitive therapeutic antibodies may be a differentiating paradigm and a novel modality for enhanced tumor targeting and improved safety profiles.
Collapse
Affiliation(s)
- Peter S Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | | | - Evan Massi
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pamela V Chew
- Oncology Biology, Gilead Sciences, Foster City, CA, USA
| | - Christine Bee
- Discovery Biology, Frontier Medicines, South San Francisco, CA, USA
| | - Padma Perkins
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Bryant Chau
- Kyverna, Synthetic Biology, Emeryville, CA, USA
| | - Kent Thudium
- Immuno-Oncology Research, Bristol Myers Squibb, Redwood City, CA, USA
| | - Jack Lohre
- In Vivo Pharmacology, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pradyot Nandi
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ekaterina G Deyanova
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Ishita Barman
- Therapeutic Discovery, 3T Biosciences, South San Francisco, CA, USA
| | - Olafur Gudmundsson
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Lawrenceville, NJ, USA
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Tim Sproul
- In Vivo Pharmacology, UNITY Biotechnology, South San Francisco, CA, USA
| | | | - Pavel Strop
- Biologics Discovery, Tallac Therapeutics, Burlingame, CA, USA
| | - Arvind Rajpal
- Large Molecule Drug Discovery, Genentech Research and Early Development, South San Francisco, CA, USA
| |
Collapse
|
103
|
Sabbatino F, Liguori L, Pepe S, Ferrone S. Immune checkpoint inhibitors for the treatment of melanoma. Expert Opin Biol Ther 2022; 22:563-576. [PMID: 35130816 PMCID: PMC9038682 DOI: 10.1080/14712598.2022.2038132] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION : Immune checkpoint inhibitor (ICI) based immunotherapy is dramatically changing the management of many types of cancers including melanoma. In this malignancy, ICIs have been shown to prolong disease and progression free survival as well as overall survival of a percentage of treated patients, becoming the cornerstone of melanoma treatment. AREAS COVERED : In this review, first, we will describe the mechanisms of immune checkpoint activation and inhibition, second, we will summarize the results obtained with ICIs in melanoma treatment in terms of efficacy as well as toxicity, third, we will discuss the potential mechanisms of immune escape from ICI, and lastly, we will review the potential predictive biomarkers of clinical efficacy of ICI-based immunotherapy in melanoma. EXPERT OPINION : ICIs represent one of the pillars of melanoma treatment. The success of ICI-based therapy is limited by the development of escape mechanisms which allow melanoma cells to avoid recognition and destruction by immune cells. These results emphasize the need of additional studies to confirm the efficacy of therapies which combine different classes of ICIs as well as ICIs with other types of therapies. Furthermore, novel and more effective predictive biomarkers are needed to better stratify melanoma patients in order to define more precisely the therapeutic algorithms.
Collapse
Affiliation(s)
- Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy 84131
| | - Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy 80131
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry 'Scuola Medica Salernitana', University of Salerno, Baronissi, Salerno, Italy 84131
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
104
|
De Silva S, Trieu H, Rajan A, Liang Y, Lin JL, Kidambi TD. Flexible sigmoidoscopy may be sufficient for initial evaluation of suspected immunotherapy-mediated colitis: A cross-sectional study. J Gastroenterol Hepatol 2022; 37:284-290. [PMID: 34547818 DOI: 10.1111/jgh.15691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/31/2021] [Accepted: 09/19/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Immune checkpoint inhibitors (ICIs) have shown promise in treating a variety of cancers. Their increased use coincides with increased incidence of immunotherapy-mediated colitis (IMC), a common adverse effect. Optimal strategy for endoscopic evaluation of IMC (full colonoscopy or flexible sigmoidoscopy) is not well-defined. METHODS Retrospective review of all patients at City of Hope referred to gastroenterology for evaluation of IMC due to gastrointestinal symptoms was performed. Patients with an existing histologic diagnosis of IMC established at an outside hospital or a diagnosis of infectious or chronic colitis were excluded. RESULTS We identified 51 symptomatic patients on ICIs prompting evaluation for IMC with colonoscopy (47/51) or flexible sigmoidoscopy (4/51). All distal rectosigmoid biopsies during flexible sigmoidoscopy demonstrated histologic evidence of IMC. In full colonoscopy, IMC was either present in all segments of colon simultaneously (35/47) or absent from all segments (12/47). No isolated proximal colonic biopsies demonstrated IMC. Endoscopically normal mucosa demonstrated histologic evidence of IMC up to 68.6% of the time. Endoscopically abnormal right, transverse, and left colon had low sensitivity (35.3%, 34.3%, and 41.7%, respectively) and high specificity (100.0%, 100.0%, and 91.7%, respectively) for histological presence of IMC. CONCLUSIONS Distal colon biopsies in patients on ICI therapy with diarrhea and suspected IMC were sufficient for diagnosing IMC in our cohort. Further, we found histologic evidence of IMC in biopsies taken from normal-appearing mucosa in a number of patients, suggesting that a normal endoscopic appearance does not preclude the presence of IMC and biopsies should be taken from both normal and abnormal-appearing mucosa.
Collapse
Affiliation(s)
- Sadie De Silva
- Division of Gastroenterology, Department of Medicine, City of Hope Medical Center, Duarte, California, USA.,Department of Medicine, Temecula Valley Hospital, Temecula, California, USA
| | - Harry Trieu
- Division of Gastroenterology, Department of Medicine, City of Hope Medical Center, Duarte, California, USA.,Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Anand Rajan
- Division of Gastroenterology, Department of Medicine, City of Hope Medical Center, Duarte, California, USA.,Department of Medicine, Olive View Medical Center, Los Angeles, California, USA
| | - Yu Liang
- Department of Pathology, City of Hope Medical Center, Duarte, California, USA
| | - James L Lin
- Division of Gastroenterology, Department of Medicine, City of Hope Medical Center, Duarte, California, USA
| | - Trilokesh D Kidambi
- Division of Gastroenterology, Department of Medicine, City of Hope Medical Center, Duarte, California, USA
| |
Collapse
|
105
|
Schwarze JK, Garaud S, Jansen YJL, Awada G, Vandersleyen V, Tijtgat J, de Wind A, Kristanto P, Seremet T, Willard-Gallo K, Neyns B. Low-Dose Nivolumab with or without Ipilimumab as Adjuvant Therapy Following the Resection of Melanoma Metastases: A Sequential Dual Cohort Phase II Clinical Trial. Cancers (Basel) 2022; 14:682. [PMID: 35158952 PMCID: PMC8833641 DOI: 10.3390/cancers14030682] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Optimal dosing and duration of adjuvant treatment with PD-1 and CTLA-4 immune checkpoint inhibitors have not been established. Prior to their regulatory approval we investigated a low-dose regimen of nivolumab with or without ipilimumab in a sequential dual-cohort phase II clinical trial. METHODS Following the complete resection of melanoma metastases, patients were treated with a single fixed dose of ipilimumab (50 mg) plus 4 bi-weekly fixed doses of nivolumab (10 mg) (cohort-1), or nivolumab for 1 year (10 mg fixed dose, Q2w x9, followed by Q8w x4) (cohort-2). Twelve-months relapse-free survival (RFS) served as the primary endpoint. RESULTS After a median follow-up of 235 weeks for cohort-1 (34 patients), and 190 weeks for cohort-2 (21 patients), the 12-months RFS-rate was, respectively, 55.9% (95% CI, 39-72), and 85.7% (95% CI, 70-100). Treatment-related adverse events occurred in 27 (79%), and 18 (86%) patients, with 3 (9%), and 1 (5%) grade 3 adverse events in cohort-1 and -2, respectively. Immunohistochemical quantification of intra- and peritumoral CD3+ T cells and CD20+ B cells, but not PD-1/PD-L1 staining, correlated significantly with RFS. CONCLUSIONS One year of adjuvant low-dose nivolumab could be an effective and economically advantageous alternative for standard dosing, at the condition of further confirmation in a larger patient cohort. A shorter low-dose nivolumab plus ipilimumab regimen seems inferior and less tolerable.
Collapse
Affiliation(s)
- Julia Katharina Schwarze
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (V.V.); (J.T.); (T.S.); (B.N.)
| | - Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Yanina J. L. Jansen
- Department of Thoracic Surgery, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (Y.J.L.J.); (K.W.-G.)
| | - Gil Awada
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (V.V.); (J.T.); (T.S.); (B.N.)
| | - Valérie Vandersleyen
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (V.V.); (J.T.); (T.S.); (B.N.)
| | - Jens Tijtgat
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (V.V.); (J.T.); (T.S.); (B.N.)
| | - Alexandre de Wind
- Department of Pathology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Paulus Kristanto
- Data Center, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Teofila Seremet
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (V.V.); (J.T.); (T.S.); (B.N.)
| | - Karen Willard-Gallo
- Department of Thoracic Surgery, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (Y.J.L.J.); (K.W.-G.)
| | - Bart Neyns
- Department of Medical Oncology, Vrije Universiteit Brussel (VUB)/Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090 Brussels, Belgium; (G.A.); (V.V.); (J.T.); (T.S.); (B.N.)
| |
Collapse
|
106
|
Maritaz C, Broutin S, Chaput N, Marabelle A, Paci A. Immune checkpoint-targeted antibodies: a room for dose and schedule optimization? J Hematol Oncol 2022; 15:6. [PMID: 35033167 PMCID: PMC8760805 DOI: 10.1186/s13045-021-01182-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-CTLA-4 and anti-PD-1/PD-L1 immune checkpoint inhibitors are therapeutic monoclonal antibodies that do not target cancer cells but are designed to reactivate or promote antitumor immunity. Dosing and scheduling of these biologics were established according to conventional drug development models, even though the determination of a maximum tolerated dose in the clinic could only be defined for anti-CTLA-4. Given the pharmacology of these monoclonal antibodies, their high interpatient pharmacokinetic variability, the actual clinical benefit as monotherapy that is observed only in a specific subset of patients, and the substantial cost of these treatments, a number of questions arise regarding the selected dose and the dosing interval. This review aims to outline the development of these immunotherapies and considers optimization options that could be used in clinical practice.
Collapse
Affiliation(s)
- Christophe Maritaz
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sophie Broutin
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Chaput
- Laboratory for Immunomonitoring in Oncology (LIO), Faculty of Pharmacy, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Unit (DITEP), LRTI U1015 INSERM, Gustave Roussy, Villejuif, France
| | - Angelo Paci
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France.
- Pharmacokinetic Unit, Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.
| |
Collapse
|
107
|
Neoadjuvant and Adjuvant Therapy for Muscle-Invasive Bladder Cancer. Urol Oncol 2022. [DOI: 10.1007/978-3-030-89891-5_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
108
|
Graziani G, Lisi L, Tentori L, Navarra P. Monoclonal Antibodies to CTLA-4 with Focus on Ipilimumab. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:295-350. [PMID: 35165868 DOI: 10.1007/978-3-030-91311-3_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The immune checkpoint cytotoxic T lymphocyte-associated antigen 4 (CTLA-4 or CD152) is a negative regulator of T-cell-mediated immune responses which plays a critical role in suppressing autoimmunity and maintaining immune homeostasis. Because of its inhibitory activity on T cells, CTLA-4 has been investigated as a drug target to induce immunostimulation, blocking the interaction with its ligands. The antitumor effects mediated by CTLA-4 blockade have been attributed to a sustained active immune response against cancer cells, due to the release of a brake on T cell activation. Ipilimumab (Yervoy, Bristol-Myers Squibb) is a fully human anti-CTLA-4 IgG1κ monoclonal antibody (mAb) that represents the first immune checkpoint inhibitor approved as monotherapy by FDA and EMA in 2011 for the treatment of unresectable/metastatic melanoma. In 2015, FDA also granted approval to ipilimumab monotherapy as adjuvant treatment of stage III melanoma to reduce the risk of tumour recurrence. The subsequent approved indications of ipilimumab for metastatic melanoma, regardless of BRAF mutational status, and other advanced/metastatic solid tumours always involve its use in association with the anti-programmed cell death protein 1 (PD-1) mAb nivolumab. Currently, ipilimumab is evaluated in ongoing clinical trials for refractory/advanced solid tumours mainly in combination with additional immunostimulating agents.
Collapse
Affiliation(s)
- Grazia Graziani
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy.
| | - Lucia Lisi
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Catholic University of the Sacred Heart, Rome, Italy
| | - Lucio Tentori
- Pharmacology Section, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Pierluigi Navarra
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
109
|
Hayes AG, Rushworth RL, Torpy DJ. Risk assessment, diagnosis, and treatment of cancer treatment-related adrenal insufficiency. Expert Rev Endocrinol Metab 2022; 17:21-33. [PMID: 34979842 DOI: 10.1080/17446651.2022.2023009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Adrenal insufficiency (AI) is an easily treatable, potentially life-threatening condition, which is increasingly recognized in malignancy. The recent introduction of immune checkpoint inhibitors, in particular, and increasing use of tyrosine kinase inhibitors have increased the frequency of AI in patients with malignancy. A review is therefore warranted to summarize current knowledge on the topic and guide safe clinical practices. AREAS COVERED Malignancy may directly impact the hypothalamic-pituitary-adrenal axis and cause AI, or their treatment including surgery, radiotherapy and medication. In this narrative review, we discuss new causes of AI, recognition of suggestive clinical features, diagnosis and subsequent treatment, aiming to avoid potentially fatal adrenal crisis (AC). Standard literature searching and authors assessment of clinical applicability were used. EXPERT OPINION Adrenal insufficiency can be easily treated once identified but life threatening if unrecognized. While use of new agents such as immune checkpoint inhibitors (ICIs) is increasing, greater understanding of the mechanism of AI is needed to target prediction tools and enhance risk stratification.
Collapse
Affiliation(s)
- Annabelle G Hayes
- Royal Adelaide Hospital, Endocrine and Metabolic Unit Adelaide, Adelaide, Australia
| | - R Louise Rushworth
- The University of Notre Dame Australia, School of Medicine, Sydney Campus Darlinghurst, Darlinghurst, Australia
| | - David J Torpy
- Royal Adelaide Hospital, Endocrine and Metabolic Unit Adelaide, Adelaide, Australia
- University of Adelaide, Discipline of Medicine, Adelaide, Australia
| |
Collapse
|
110
|
Tian M, Zhang S, Tseng Y, Shen X, Dong L, Xue R. Gut Microbiota and Immune Checkpoint Inhibitors-Based Immunotherapy. Anticancer Agents Med Chem 2022; 22:1244-1256. [PMID: 34229597 DOI: 10.2174/1871520621666210706110713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/15/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
Application of immune checkpoint inhibitors (ICIs) is a major breakthrough in the field of cancer therapy, which has displayed tremendous potential in various types of malignancies. However, their response rates range widely in different cancer types and a significant number of patients experience immune-related adverse effects (irAEs) induced by these drugs, limiting the proportion of patients who can truly benefit from ICIs. Gut microbiota has gained increasing attention due to its emerging role in regulating the immune system. In recent years, numerous studies have shown that gut microbiota can modulate antitumor response, as well as decrease the risk of colitis due to ICIs in patients receiving immunotherapy. The present review analyzed recent progress of relevant basic and clinical studies in this area and explored new perspectives to enhance the efficacy of ICIs and alleviate associated irAEs via manipulation of the gut microbiota.
Collapse
Affiliation(s)
- Mingming Tian
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Si Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Glycoconjugate Research Ministry of Public Health, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yujen Tseng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200032,China
| | - Xizhong Shen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital of Fudan University, Shanghai, 200032, China
| |
Collapse
|
111
|
Korman AJ, Garrett-Thomson SC, Lonberg N. The foundations of immune checkpoint blockade and the ipilimumab approval decennial. Nat Rev Drug Discov 2021; 21:509-528. [PMID: 34937915 DOI: 10.1038/s41573-021-00345-8] [Citation(s) in RCA: 323] [Impact Index Per Article: 80.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2021] [Indexed: 12/11/2022]
Abstract
Cancer immunity, and the potential for cancer immunotherapy, have been topics of scientific discussion and experimentation for over a hundred years. Several successful cancer immunotherapies - such as IL-2 and interferon-α (IFNα) - have appeared over the past 30 years. However, it is only in the past decade that immunotherapy has made a broad impact on patient survival in multiple high-incidence cancer indications. The emergence of immunotherapy as a new pillar of cancer treatment (adding to surgery, radiation, chemotherapy and targeted therapies) is due to the success of immune checkpoint blockade (ICB) drugs, the first of which - ipilimumab - was approved in 2011. ICB drugs block receptors and ligands involved in pathways that attenuate T cell activation - such as cytotoxic T lymphocyte antigen 4 (CTLA4), programmed cell death 1 (PD1) and its ligand, PDL1 - and prevent, or reverse, acquired peripheral tolerance to tumour antigens. In this Review we mark the tenth anniversary of the approval of ipilimumab and discuss the foundational scientific history of ICB, together with the history of the discovery, development and elucidation of the mechanism of action of the first generation of drugs targeting the CTLA4 and PD1 pathways.
Collapse
|
112
|
Optimized Dosing: The Next Step in Precision Medicine in Non-Small-Cell Lung Cancer. Drugs 2021; 82:15-32. [PMID: 34894338 DOI: 10.1007/s40265-021-01654-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
In oncology, and especially in the treatment of non-small-cell lung cancer (NSCLC), dose optimization is often a neglected part of precision medicine. Many drugs are still being administered in "one dose fits all" regimens or based on parameters that are often only minor determinants for systemic exposure. These dosing approaches often introduce additional pharmacokinetic variability and do not add to treatment outcomes. Fortunately, pharmacological knowledge is increasing, providing valuable information regarding the potential of, for example, therapeutic drug monitoring. This article focuses on the evidence for the most promising and easily implemented optimized dosing approaches for the small-molecule inhibitors, chemotherapeutic agents, and monoclonal antibodies as treatment options currently approved for NSCLC. Despite limitations such as investigations having been conducted in oncological diseases other than NSCLC or the retrospective origin of many analyses, an alternative dosing regimen could be beneficial for treatment outcomes, prescriber convenience, or financial burden on healthcare systems. This review of the literature provides recommendations on the implementation of dose optimization and advice regarding promising strategies that deserve further research in NSCLC.
Collapse
|
113
|
Filleron T, Bachelier M, Mazieres J, Pérol M, Meyer N, Martin E, Mathevet F, Dauxois JY, Porcher R, Delord JP. Assessment of Treatment Effects and Long-term Benefits in Immune Checkpoint Inhibitor Trials Using the Flexible Parametric Cure Model: A Systematic Review. JAMA Netw Open 2021; 4:e2139573. [PMID: 34932105 PMCID: PMC8693223 DOI: 10.1001/jamanetworkopen.2021.39573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPORTANCE Compared with standard cytotoxic therapies, randomized immune checkpoint inhibitor (ICI) phase 3 trials reveal delayed benefits in terms of patient survival and/or long-term response. Such outcomes generally violate the assumption of proportional hazards, and the classical Cox proportional hazards regression model is therefore unsuitable for these types of analyses. OBJECTIVE To evaluate the ability of the flexible parametric cure model (FPCM) to estimate treatment effects and long-term responder fractions (LRFs) independently of prespecified time points. EVIDENCE REVIEW This systematic review used reconstructed individual patient data from ICI advanced or metastatic melanoma and lung cancer phase 3 trials extracted from the literature. Trials published between January 1, 2010, and October 1, 2019, with long-term follow-up periods (maximum follow-up, ≥36 months in first line and ≥30 months otherwise) were selected to identify LRFs. Individual patient data for progression-free survival were reconstructed from the published randomized ICI phase 3 trial results. The FPCM was applied to estimate treatment effects on the overall population and on the following components of the population: LRF and progression-free survival in non-long-term responders. Results obtained were compared with treatment effects estimated using the Cox proportional hazards regression model. FINDINGS In this systematic review, among the 23 comparisons studied using the FPCM, a statistically significant association between the time-to-event component and experimental treatment was observed in the main analyses and confirmed in the sensitivity analyses of 18 comparisons. Results were discordant for 4 comparisons that were not significant by the Cox proportional hazards regression model. The LRFs varied from 1.5% to 12.7% for the control arms and from 4.6% to 38.8% for the experimental arms. Differences in LRFs varied from 2% to 29% and were significantly increased in the experimental compared with the control arms, except for 4 comparisons. CONCLUSIONS AND RELEVANCE This systematic review of reconstructed individual patient data found that the FPCM was a complementary approach that provided a comprehensive and pertinent evaluation of benefit and risk by assessing whether ICI treatment was associated with an increased probability of patients being long-term responders or with an improved progression-free survival in patients who were not long-term responders.
Collapse
Affiliation(s)
- Thomas Filleron
- Department of Biostatistics, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse, Toulouse, France
| | - Marine Bachelier
- Department of Biostatistics, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse, Toulouse, France
| | - Julien Mazieres
- Department of Pneumology, Centre Hospitalier Universitaire de Toulouse Larrey, Toulouse, France
| | - Maurice Pérol
- Department of Medical Oncology, Léon Bérard Cancer Center, Lyon, France
| | - Nicolas Meyer
- Institut Universitaire du Cancer Toulouse Oncopôle, Toulouse, France
| | - Elodie Martin
- Department of Biostatistics, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse, Toulouse, France
| | - Fanny Mathevet
- Department of Biostatistics, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse, Toulouse, France
| | - Jean-Yves Dauxois
- Institut de Mathématiques de Toulouse, Université de Toulouse, Centre National de la Recherche Scientifique, Institut National des Sciences Appliquées de Toulouse, Toulouse, France
| | - Raphael Porcher
- Assistance Publique des Hôpitaux de Paris, Hôpital Hôtel Dieu, Centre d’Épidémiologie Clinique, INSERM U1153, Paris, France
| | - Jean-Pierre Delord
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse, Toulouse, France
| |
Collapse
|
114
|
Davis L, Tarduno A, Lu YC. Neoantigen-Reactive T Cells: The Driving Force behind Successful Melanoma Immunotherapy. Cancers (Basel) 2021; 13:cancers13236061. [PMID: 34885172 PMCID: PMC8657037 DOI: 10.3390/cancers13236061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Cancer immunotherapy is a revolutionary type of cancer therapy. It uses the patient’s own immune system to fight and potentially cure cancer. The first major breakthrough of immunotherapy came from successful clinical trials for melanoma treatments. Since then, researchers have focused on understanding the science behind immunotherapy, so that patients with other types of cancer may also benefit. One of the major findings is that the T cells in melanoma patients may recognize a specific type of tumor antigen, called neoantigens, and then kill tumor cells that present these neoantigens. The neoantigens mainly arise from the DNA mutations found in tumor cells. These mutations are translated into mutated proteins that are then distinguished by T cells. In this article, we discuss the critical role of T cells in immunotherapy, as well as the clinical trials that shaped the treatments for melanoma. Abstract Patients with metastatic cutaneous melanoma have experienced significant clinical responses after checkpoint blockade immunotherapy or adoptive cell therapy. Neoantigens are mutated proteins that arise from tumor-specific mutations. It is hypothesized that the neoantigen recognition by T cells is the critical step for T-cell-mediated anti-tumor responses and subsequent tumor regressions. In addition to describing neoantigens, we review the sentinel and ongoing clinical trials that are helping to shape the current treatments for patients with cutaneous melanoma. We also present the existing evidence that establishes the correlations between neoantigen-reactive T cells and clinical responses in melanoma immunotherapy.
Collapse
Affiliation(s)
- Lindy Davis
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Ashley Tarduno
- Department of Surgery, Albany Medical Center, Albany, NY 12208, USA; (L.D.); (A.T.)
| | - Yong-Chen Lu
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Correspondence:
| |
Collapse
|
115
|
Cai X, Zhan H, Ye Y, Yang J, Zhang M, Li J, Zhuang Y. Current Progress and Future Perspectives of Immune Checkpoint in Cancer and Infectious Diseases. Front Genet 2021; 12:785153. [PMID: 34917131 PMCID: PMC8670224 DOI: 10.3389/fgene.2021.785153] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
The inhibitory regulators, known as immune checkpoints, prevent overreaction of the immune system, avoid normal tissue damage, and maintain immune homeostasis during the antimicrobial or antiviral immune response. Unfortunately, cancer cells can mimic the ligands of immune checkpoints to evade immune surveillance. Application of immune checkpoint blockade can help dampen the ligands expressed on cancer cells, reverse the exhaustion status of effector T cells, and reinvigorate the antitumor function. Here, we briefly introduce the structure, expression, signaling pathway, and targeted drugs of several inhibitory immune checkpoints (PD-1/PD-L1, CTLA-4, TIM-3, LAG-3, VISTA, and IDO1). And we summarize the application of immune checkpoint inhibitors in tumors, such as single agent and combination therapy and adverse reactions. At the same time, we further discussed the correlation between immune checkpoints and microorganisms and the role of immune checkpoints in microbial-infection diseases. This review focused on the current knowledge about the role of the immune checkpoints will help in applying immune checkpoints for clinical therapy of cancer and other diseases.
Collapse
Affiliation(s)
- Xin Cai
- Heilongjiang Administration of Traditional Chinese Medicine, Harbin, China
| | - Huajie Zhan
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Yuguang Ye
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jinjin Yang
- Department of Pathology, Harbin Medical University, Harbin, China
| | - Minghui Zhang
- Department of Oncology, Chifeng City Hospital, Chifeng, China
- *Correspondence: Yuan Zhuang, ; Jing Li, ; Minghui Zhang,
| | - Jing Li
- Department of Pathology and Electron Microscopy Center, Harbin Medical University, Harbin, China
- *Correspondence: Yuan Zhuang, ; Jing Li, ; Minghui Zhang,
| | - Yuan Zhuang
- Department of Pathology, Harbin Medical University, Harbin, China
- *Correspondence: Yuan Zhuang, ; Jing Li, ; Minghui Zhang,
| |
Collapse
|
116
|
Liu X, Shi Y, Zhang D, Zhou Q, Liu J, Chen M, Xu Y, Zhao J, Zhong W, Wang M. Risk factors for immune-related adverse events: what have we learned and what lies ahead? Biomark Res 2021; 9:79. [PMID: 34732257 PMCID: PMC8565046 DOI: 10.1186/s40364-021-00314-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/08/2021] [Indexed: 12/18/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have heralded the advent of a new era in oncology by holding the promise of prolonged survival in severe and otherwise treatment-refractory advanced cancers. However, the remarkable antitumor efficacy of these agents is overshadowed by their potential for inducing autoimmune toxic effects, collectively termed immune-related adverse events (irAEs). These autoimmune adverse effects are often difficult to predict, possibly permanent, and occasionally fatal. Hence, the identification of risk factors for irAEs is urgently needed to allow for prompt therapeutic intervention. This review discusses the potential mechanisms through which irAEs arise and summarizes the existing evidence regarding risk factors associated with the occurrence of irAEs. In particular, we examined available data regarding the effect of a series of clinicopathological and demographic factors on the risk of irAEs.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Yuequan Shi
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Dongming Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Jia Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China
| | - Wei Zhong
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China.
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No 1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China.
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China.
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No 1 Shuaifuyuan Wangfujing, Dongcheng District, 100730, Beijing, China.
| |
Collapse
|
117
|
Gil-Rojas Y, Lasalvia P, Hernández F, Castañeda-Cardona C, Castrillón-Correa J, Herrera D, Rosselli D. Cost-Effectiveness of the Dabrafenib Schedule in Combination With Trametinib Compared With Other Targeted Therapies, Immunotherapy, and Dacarbazine for the Treatment of Unresectable or Metastatic Melanoma With BRAFV600 Mutation in Colombia. Value Health Reg Issues 2021; 26:182-190. [PMID: 34673349 DOI: 10.1016/j.vhri.2021.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Advanced melanoma accounts for 4% of malignant skin tumors, and approximately 80% of deaths are attributed to it. The most frequent mutation of the RAF gene is BRAFV600, which has been associated with a worse prognosis. The objective of the research was to evaluate the cost-effectiveness of the combined regimen of dabrafenib plus trametinib (D + T) compared with other targeted therapies, immunotherapy, and dacarbazine for the treatment of unresectable/metastatic melanoma with BRAFV600 mutation from the perspective of the Colombian health system. METHODS A partitioned survival model with 3 states (progression-free survival, progression, and death) was used to evaluate the cost-effectiveness for a time horizon of 20 years. Owing to the perspective of the analysis, only direct medical costs were taken into account. The efficacy of the evaluated treatment and the comparators were measured in terms of overall survival and progression-free survival. All costs were expressed in Colombian pesos as of 2018, and outcomes and costs were discounted at 5% annually. Two analysis scenarios were considered, one in which only monitoring and follow-up costs were included in the progression phase and another in which costs of acquisition of possible treatment sequences were also included. RESULTS In the first scenario (without postprogression medication costs), the combined D + T regimen was a dominant alternative to vemurafenib + cobimetinib but was not a cost-effective option compared with vemurafenib, nivolumab, ipilimumab, nivolumab + ipilimumab, pembrolizumab, and dacarbazine. In the second scenario (with drug costs in postprogression), D + T was dominant compared with vemurafenib + cobimetinib and cost-effective compared with nivolumab and pembrolizumab. Compared with other schemes, the incremental cost-effectiveness ratio was above the threshold of 3 gross domestic product per capita. Probabilistic sensitivity analyses showed that a willingness-to-pay threshold of Col$56 484 300 (US$19 108) per quality-adjusted life-year would not be reached at the current price of schema in Colombia. CONCLUSIONS The combined scheme could be a cost-effective and even a cost-saving alternative to vemurafenib + cobimetinib, nivolumab, and pembrolizumab if the costs associated with the use of other medications are taken into account after progression to the first line of treatment. Compared with the other comparators, it produces a greater number of quality-adjusted life-years, but the incremental cost-effectiveness ratio is above that of the willingness to pay.
Collapse
Affiliation(s)
- Yaneth Gil-Rojas
- Department of Economic Studies, Neuroeconomix, Bogotá, Colombia.
| | | | | | | | | | | | - Diego Rosselli
- Faculty of Medicine, Department of Clinical Epidemiology and Biostatistics, Pontificia Universidad Javeriana, Hospital San Ignacio, Bogotá, Colombia
| |
Collapse
|
118
|
Long GV, Robert C, Butler MO, Couture F, Carlino MS, O'Day S, Atkinson V, Cebon JS, Brown MP, Dalle S, Hill AG, Gibney GT, McCune S, Menzies AM, Niu C, Ibrahim N, Moreno BH, Diab A. Standard-Dose Pembrolizumab Plus Alternate-Dose Ipilimumab in Advanced Melanoma: KEYNOTE-029 Cohort 1C, a Phase 2 Randomized Study of Two Dosing Schedules. Clin Cancer Res 2021; 27:5280-5288. [PMID: 34210681 PMCID: PMC9401495 DOI: 10.1158/1078-0432.ccr-21-0793] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/16/2021] [Accepted: 06/25/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Standard-dose pembrolizumab plus alternative-dose ipilimumab (1 mg/kg Q3W for 4 doses) were tolerable and had robust antitumor activity in advanced melanoma in cohort B of the phase 1 KEYNOTE-029 study. Cohort C evaluated standard-dose pembrolizumab with two other alternative ipilimumab regimens. PATIENTS AND METHODS Patients with treatment-naive unresectable stage III/IV melanoma were randomly assigned 1:1 to pembrolizumab 200 mg Q3W for ≤24 months plus ipilimumab 50 mg Q6W for 4 doses (PEM200+IPI50), or the same pembrolizumab regimen plus ipilimumab 100 mg Q12W for 4 doses (PEM200+IPI100). Primary end points were incidence of grade 3-5 treatment-related adverse events (TRAE) and objective response rate (ORR) per RECIST v1.1 by independent central review. Per protocol-defined thresholds, grade 3-5 TRAE incidence ≤26% indicated meaningful toxicity reduction and ORR ≥48% indicated no decrease in efficacy versus data reported for other PD-1 inhibitor/ipilimumab combinations. RESULTS Median follow-up on February 18, 2019, was 16.3 months in PEM200+IPI50 (N = 51) and 16.4 months in PEM200+IPI100 (N = 51). Grade 3-5 TRAEs occurred in 12 (24%) patients in PEM200+IPI50 and 20 (39%) in PEM200+IPI100. One patient in PEM200+IPI50 died from treatment-related autoimmune myocarditis. Immune-mediated AEs or infusion reactions occurred in 21 (42%) patients in PEM200+IPI50 and 28 (55%) in PEM200+IPI100. ORR was 55% in PEM200+IPI50; 61% in PEM200+IPI100. CONCLUSIONS Pembrolizumab 200 mg Q3W plus ipilimumab 50 mg Q6W or 100 mg Q12W demonstrated antitumor activity above the predefined threshold; pembrolizumab plus ipilimumab 50 mg Q6W had lower incidence of grade 3-5 TRAEs than the predefined threshold, suggesting a reduction in toxicity. See related commentary by Jameson-Lee and Luke, p. 5153.
Collapse
Affiliation(s)
- Georgina V. Long
- Department of Medical Oncology and Translational Research, Melanoma Institute Australia, The University of Sydney, and Mater and Royal North Shore Hospitals, Sydney, New South Wales, Australia
| | - Caroline Robert
- Department of Medicine, Dermatology Service, Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Marcus O. Butler
- Department of Medical Oncology, The Princess Margaret Cancer Center and University of Toronto, Toronto, Ontario, Canada
| | - Felix Couture
- Department of Hematology, CHU de Québec—Hôtel-Dieu de Québec, Québec City, Québec, Canada
| | - Matteo S. Carlino
- Department of Medicine, Westmead and Blacktown Hospitals, Melanoma Institute Australia, and The University of Sydney, Sydney, New South Wales, Australia
| | - Steven O'Day
- Department of Medical Oncology, John Wayne Cancer Institute at Providence Saint John's Health Center, Santa Monica, California
| | - Victoria Atkinson
- University of Queensland, Division of Cancer Sciences, Gallipoli Medical Research Foundation, Greenslopes Private Hospital, Brisbane, Queensland, Australia
| | - Jonathan S. Cebon
- Department of Hematology/Oncology, Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
| | - Michael P. Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital and University of Adelaide, Adelaide, South Australia, Australia
| | - Stéphane Dalle
- University of Lyon, Hospices Civils de Lyon, Cancer Research Center of Lyon, Lyon, France
| | - Andrew G. Hill
- Department of Health, Tasman Oncology Research, Southport, Queensland, Australia
| | - Geoffrey T. Gibney
- Melanoma Disease Group, Georgetown-Lombardi Comprehensive Cancer Center, Washington, DC
| | - Steven McCune
- Department of Clinical Research, Wellstar Health System, Marietta, Georgia
| | - Alexander M. Menzies
- Department of Medical Oncology and Translational Research, Melanoma Institute Australia, The University of Sydney, and Mater and Royal North Shore Hospitals, Sydney, New South Wales, Australia
| | - Cuizhen Niu
- Department of Clinical Oncology, MSD China, Beijing, China
| | - Nageatte Ibrahim
- Department of Clinical Oncology, Merck & Co., Inc., Kenilworth, New Jersey
| | | | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
119
|
HCC and Molecular Targeting Therapies: Back to the Future. Biomedicines 2021; 9:biomedicines9101345. [PMID: 34680462 PMCID: PMC8533575 DOI: 10.3390/biomedicines9101345] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of death from cancer in the world. Recently, the effectiveness of new antiviral therapies and the HBV vaccine have reduced HCC’s incidence, while non-alcoholic steato-hepatitis is an emerging risk factor. This review focuses on antiangiogenic molecules and immune checkpoint inhibitors approved for HCC treatment and possible future approaches. Sorafenib was the first drug approved for the treatment of advanced HCC (aHCC) and it has been shown to increase survival by a few months. Lenvatinib, a multikinase inhibitor, has shown non-inferiority in survival compared with sorafenib and an improvement in progression-free survival (PFS). The combination of atezolizumab (an anti-PDL1 antibody) and bevacizumab (an anti-VEGF antibody) was the first drug combination approved for HCC, demonstrating improved survival compared with sorafenib (19.2 vs. 13.4 months). As a second line of therapy, three regimens (regorafenib, cabozantinib, and ramucirumab) have been approved for the treatment of aHCC after progression on sorafenib according to guidelines. Furthermore, nivolumab, pembrolizumab, and nivolumab plus ipilimumab have been approved by the FDA (2017, 2018, and 2020, respectively). Finally, immune target therapy, cancer vaccines, and epigenetic drugs represent three new possible weapons for the treatment of HCC.
Collapse
|
120
|
Yuan J, Khilnani A, Brody J, Andtbacka RHI, Hu-Lieskovan S, Luke JJ, Diab A, Marabelle A, Snyder A, Cao ZA, Hodi FS. Current strategies for intratumoural immunotherapy - Beyond immune checkpoint inhibition. Eur J Cancer 2021; 157:493-510. [PMID: 34561127 DOI: 10.1016/j.ejca.2021.08.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
Immunotherapy has revolutionised cancer treatment through restoration of host antitumour immune response. Immune checkpoint inhibitors (ICIs) confer durable responses in only a subset of patients. Mechanisms of ICI resistance to improve durable response rates and overall survival are an area of intense clinical research. Robust clinical development is ongoing to evaluate novel combination therapies to overcome ICI resistance, including targeting immunoregulatory pathways in the tumour microenvironment. Intratumoural (IT) immunotherapies such as toll-like receptor agonists, stimulator of interferon-induced gene agonists, retinoic-inducible gene I-like receptor agonists and oncolytic viruses may represent potential combination treatment options to overcome ICI resistance. Use of IT immunotherapies in combination with ICIs may alter the tumour microenvironment to address resistance mechanisms and improve antitumour response. Optimisation of IT immunotherapy clinical trials will elucidate resistance mechanisms, facilitate clinical trial design, define pharmacodynamic predictors that identify patients who may most benefit and inform clinical development of combination immunotherapy regimens. Here we provide an overview of IT immunotherapy principles, mechanisms of action, categories of IT immunotherapeutics, emerging data, clinical development strategies, response assessment, dose and schedule determination, clinical trial design and translational study design.
Collapse
Affiliation(s)
- Jianda Yuan
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - Anuradha Khilnani
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - Joshua Brody
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| | - Robert H I Andtbacka
- Seven and Eight Biopharmaceuticals Inc., 343 Thornall Street, Suite 520, Edison, NJ, 08837, USA.
| | - Siwen Hu-Lieskovan
- Department of Internal Medicine-Oncology, Huntsman Cancer Institute, University of Utah, 2000 Cir of Hope Dr #1950, Salt Lake City, UT, 84112, USA.
| | - Jason J Luke
- Department of Hematology/Oncology, University of Pittsburgh Hillman Cancer Center, 5115 Centre Ave, Pittsburgh, PA, 15232, USA.
| | - Adi Diab
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| | - Aurelien Marabelle
- Department of Therapeutic Innovation and Early Trials, Gustave Roussy, University of Paris-Saclay, 114 Rue Edouard Vaillant, 94805 Villejuif, France.
| | - Alexandra Snyder
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - Z Alexander Cao
- Department of Translational Oncology and Early Oncology Development, Merck & Co., Inc., 2000 Galloping Hill Rd, Kenilworth, NJ, 07033, USA.
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA.
| |
Collapse
|
121
|
Gonzalez BD, Eisel SL, Bowles KE, Hoogland AI, James BW, Small BJ, Sharpe S, Hyland KA, Bulls HW, Christy SM, Mansfield J, Nelson AM, Alla R, Maharaj K, Kennedy B, Lafranchise E, Williams NL, Jennewein S, Oswald LB, Postow MA, Dicker AP, Jim HSL. Meta-Analysis of Quality of Life in Cancer Patients Treated with Immune Checkpoint Inhibitors. J Natl Cancer Inst 2021; 114:808-818. [PMID: 34508604 DOI: 10.1093/jnci/djab171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/17/2021] [Accepted: 08/24/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Trials of immune checkpoint inhibitors (ICIs) have published patient-reported quality of life (QOL), but the size and heterogeneity of this literature can make patient education difficult. This meta-analysis aimed to describe change in QOL and symptomatology in patients receiving ICIs for cancer. METHODS Following PRISMA guidelines, databases were searched through November 2019 for articles or abstracts of prospective, original studies reporting longitudinal QOL in adult cancer patients treated with ICIs. The prespecified primary outcomes were change in global QOL among patients treated with ICIs and difference in change since baseline in global QOL between patients treated with ICI vs. non-ICI active treatment. Secondary outcomes included physical functioning and symptomatology. All statistical tests were 2-sided. RESULTS Twenty-six of 20,323 publications met inclusion criteria. Global QOL did not change over time in patients treated with ICIs (k = 26, n = 6,974, P = .19). Larger improvements in global QOL was observed in patients receiving ICI vs. non-ICI regimens (k = 16, ICI n = 3,588, non-ICI n = 2,948, P < .001). Physical functioning did not change in patients treated with ICIs (k = 14, n = 3,169, P=.47); there were no differences in mean change between ICI vs. non-ICI regimens (k = 11, n = 4,630, P=.94. Regarding symptoms, appetite loss, insomnia, and pain severity decreased but dyspnea severity increased in patients treated with ICIs (k = 14, n = 3,243-3,499) (Ps < 0.001). Insomnia severity was higher in patients treated with ICIs than non-ICI regimens (k = 11, n = 4,791) (P < .001). CONCLUSIONS This study is among the first to quantitatively summarize QOL in patients treated with ICIs. Findings suggest ICI recipients report no change in global QOL and higher QOL than patients treated with non-ICI regimens.
Collapse
Affiliation(s)
- Brian D Gonzalez
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Sarah L Eisel
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Kristina E Bowles
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Aasha I Hoogland
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Brian W James
- Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Brent J Small
- School of Aging Studies, University of South Florida, Tampa, FL
| | - Susan Sharpe
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Kelly A Hyland
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Hailey W Bulls
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Shannon M Christy
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Jori Mansfield
- Morsani College of Medicine, University of South Florida, Tampa, FL
| | | | - Raviteja Alla
- School of Aging Studies, University of South Florida, Tampa, FL
| | - Kelly Maharaj
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Brittany Kennedy
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | | | | | - Sarah Jennewein
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Laura B Oswald
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| | - Michael A Postow
- Southeast Radiation Oncology Group, Levine Cancer Institute at Atrium Health, Charlotte, North Carolina
| | | | - Heather S L Jim
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
122
|
Abstract
INTRODUCTION Checkpoint inhibitor drugs including ipilimumab have been reported to induce intestinal injury. OBJECTIVE We aimed to evaluate the risk of chronic (> 6 weeks) enterocolitis following ipilimumab administration, and the likelihood that an enteritis vs colitis or enterocolitis is seen. PATIENTS AND METHODS We searched MEDLINE, EMBASE, CENTRAL, the World Health Organization International Clinical Trials Registry, and conference proceedings. We included: (1) randomized controlled trials comparing ipilimumab administration with placebo/standard care/other active chemotherapy regimens and (2) prospective observational studies. Separate meta-analyses were performed for randomized controlled trials and observational studies. RESULTS Of 4760 records, we included ten unique randomized controlled trials (n = 5814 subjects) and 34 unique prospective observational studies (n = 3699 subjects). In randomized controlled trials, the pooled relative risk of ≥ grade 3 enterocolitis or ≥ grade 3 diarrhea associated with ipilimumab was 13.31 (95% confidence interval 6.01-29.48, I2 = 0%, ten trials) and 6.72 (95% confidence interval 3.30-13.65, I2 = 63%, ten trials), respectively. In observational studies, the 3-monthly risk of developing grade 3 or higher enteritis, colitis, or enterocolitis was 4% (95% confidence interval 3-7, I2 = 77.40%, 25 studies). Randomized controlled trials and observational studies did not distinguish between acute and chronic enterocolitis. Of the included observational studies, the pooled risk of incurring small bowel involvement associated with ipilimumab was 1% (95% CI 0-4, I2 = 0%, four studies) per every 3-month time period. CONCLUSIONS Insufficient data exist to quantify or distinguish the risk of acute vs chronic enterocolitis following ipilmumab use. Because of the serious impact of chronic enterocolitis on quality of life and further cancer treatment, future trials evaluating the safety of immunotherapy should report gastrointestinal events in greater detail.
Collapse
|
123
|
Nienhuis PH, Antunes IF, Glaudemans AWJM, Jalving M, Leung D, Noordzij W, Slart RHJA, de Vries EF, Hospers GAP. 18F-BMS986192 PET imaging of PD-L1 in metastatic melanoma patients with brain metastases treated with immune checkpoint inhibitors. A pilot study. J Nucl Med 2021; 63:899-905. [PMID: 34503964 DOI: 10.2967/jnumed.121.262368] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) targeting PD-1/PD-L1 frequently induces tumor response in metastatic melanoma patients. However, tumor response often takes months and may be heterogeneous. Consequently, additional local treatment for non-responsive metastases may be needed, especially in the case of brain metastases. Non-invasive imaging may allow the characterization of (brain) metastases to predict response. This pilot study uses 18F-BMS986192 PET for PD-L1 expression to explore the variability in metastatic tracer uptake and its relation to tumor response, with a special focus on brain metastases. Methods: Metastatic melanoma patients underwent whole-body 18F-BMS986192 PET/CT scanning before and 6 weeks after starting ICI therapy. 18F-BMS986192 uptake was measured in healthy tissues, organs, and tumor lesions. Tumor response was evaluated at 12 weeks using CT thorax/abdomen and MRI brain. RECIST v 1.1 was used to define therapy response per patient. Response per lesion was measured by the percentage change in lesion diameter. Toxicity was assessed according to Common Terminology Criteria for Adverse Events version 4.0. Results: Baseline 18F-BMS986192 PET/CT was performed in 8 patients, with follow-up scans in 4 patients. The highest tracer uptake was observed in the spleen, bone marrow, kidneys, and liver. Tracer uptake in tumor lesions was heterogeneous. In total, 42 tumor lesions were identified at baseline with most lesions in the lungs (n = 21) and brain (n = 14). Tracer uptake was similar between tumor locations. 18F-BMS986192 uptake in lesions at baseline, corrected for blood pool activity, was negatively correlated with the change lesion diameter at response evaluation (r=-0.49, P = 0.005), both in intra- and extracerebral lesions. Receiver operating characteristic (ROC) analysis demonstrated that 18F-BMS986192 uptake can discriminate between responding and nonresponding lesions with an area under the curve of 0.82. At the follow-up scan an increased 18F-BMS986192 uptake compared to baseline scan was correlated with an increased lesion diameter at response evaluation. In the follow-up 18F-BMS986192-PET scan of two patients, ICI-related toxicity (thyroiditis and colitis) was detected. Conclusion: In this pilot study, 18F-BMS986192 PET showed heterogeneous uptake in intra and extracerebral metastatic lesions in melanoma patients. Baseline 18F-BMS986192 uptake was able to predict an ICI treatment-induced reduction in lesion volume, whereas the follow-up PET scan allowed the detection of treatment-induced toxicity.
Collapse
|
124
|
Melero I, Castanon E, Alvarez M, Champiat S, Marabelle A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat Rev Clin Oncol 2021; 18:558-576. [PMID: 34006998 PMCID: PMC8130796 DOI: 10.1038/s41571-021-00507-y] [Citation(s) in RCA: 300] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2021] [Indexed: 02/04/2023]
Abstract
Immune-checkpoint inhibitors and chimeric antigen receptor (CAR) T cells are revolutionizing oncology and haematology practice. With these and other immunotherapies, however, systemic biodistribution raises safety issues, potentially requiring the use of suboptimal doses or even precluding their clinical development. Delivering or attracting immune cells or immunomodulatory factors directly to the tumour and/or draining lymph nodes might overcome these problems. Hence, intratumoural delivery and tumour tissue-targeted compounds are attractive options to increase the in situ bioavailability and, thus, the efficacy of immunotherapies. In mouse models, intratumoural administration of immunostimulatory monoclonal antibodies, pattern recognition receptor agonists, genetically engineered viruses, bacteria, cytokines or immune cells can exert powerful effects not only against the injected tumours but also often against uninjected lesions (abscopal or anenestic effects). Alternatively, or additionally, biotechnology strategies are being used to achieve higher functional concentrations of immune mediators in tumour tissues, either by targeting locally overexpressed moieties or engineering 'unmaskable' agents to be activated by elements enriched within tumour tissues. Clinical trials evaluating these strategies are ongoing, but their development faces issues relating to the administration methodology, pharmacokinetic parameters, pharmacodynamic end points, and immunobiological and clinical response assessments. Herein, we discuss these approaches in the context of their historical development and describe the current landscape of intratumoural or tumour tissue-targeted immunotherapies.
Collapse
Affiliation(s)
- Ignacio Melero
- Department of Immunology, Clínica Universidad de Navarra, Pamplona, Spain.
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
- Program for Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Eduardo Castanon
- Department of Immunology, Clínica Universidad de Navarra, Pamplona, Spain
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Maite Alvarez
- Program for Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Université Paris Saclay, Gustave Roussy, Villejuif, France
- INSERM U1015, Gustave Roussy, Villejuif, France
- Biotherapies for In Situ Antitumor Immunization (BIOTHERIS), Centre d'Investigation Clinique INSERM CICBT1428, Villejuif, France
| | - Aurelien Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Université Paris Saclay, Gustave Roussy, Villejuif, France.
- INSERM U1015, Gustave Roussy, Villejuif, France.
- Biotherapies for In Situ Antitumor Immunization (BIOTHERIS), Centre d'Investigation Clinique INSERM CICBT1428, Villejuif, France.
| |
Collapse
|
125
|
Ge Y, Zhang H, Weygant N, Yao J. Differential Dermatologic Adverse Events Associated With Checkpoint Inhibitor Monotherapy and Combination Therapy: A Meta-Analysis of Randomized Control Trials. Front Pharmacol 2021; 12:640099. [PMID: 34447305 PMCID: PMC8383780 DOI: 10.3389/fphar.2021.640099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background: As immune checkpoint inhibitors (ICIs) transition to the forefront of cancer treatment, a better understanding of immune related adverse events (IRAEs) is essential to promote safe clinical practice. Dermatologic adverse events are the most common IRAEs and can lead to drug withdrawal and decreased quality of life. This meta-analysis aimed to investigate the risk of the most prevalent dermatologic adverse events (pruritus and rash) among various ICI treatment regimens. Methods: A systematic search of electronic databases was performed to identify qualified randomized controlled trials (RCTs). Data for any grade and high grade pruritus and rash were extracted for meta-analysis. Two reviewers independently assessed methodological quality. The relative risk summary and 95% confidence interval were calculated. Results: 50 RCTs involving 29941 patients were analyzed. The risk of pruritus (2.15 and 4.21 relative risk respectively) and rash (1.61 and 3.89 relative risk respectively) developing from CTLA-4 or PD-1/-L1 inhibitor were increased compared to placebo, but this effect was not dose-dependent. PD-1/-L1 plus CTLA-4 inhibitor was associated with increased risk of pruritus (1.76 and 0.98 relative risk respectively) and rash (1.72 and 1.37 relative risk respectively) compared to either monotherapy. Compared with CTLA-4 inhibitor, PD-1/-L1 inhibitor had a significantly decreased risk of pruritus and rash in both monotherapy and combination therapy (0.65 and 0.29 relative risk respectively). No significant difference was found between PD-1/-L1 inhibitor combined with chemotherapy and PD-1/-L1 monotherapy in any grade and high grade rash (0.84 and 1.43 relative risk respectively). In subgroup analyses, PD-1 inhibitor was associated with reduced risk of pruritus and rash compared to PD-L1 inhibitor. Conclusion: Our meta-analysis demonstrates a better safety profile for PD-1/-L1 inhibitor compared to CTLA-4 inhibitor in terms of pruritus and rash among both monotherapy and multiple combination therapies. PD-L1 inhibitor may contribute to an increased risk of pruritus and rash compared to PD-1 inhibitor.
Collapse
Affiliation(s)
- Yang Ge
- Beijing Chao-Yang Hospital, Dept. of Oncology, Capital Medical University, Beijing, China
| | - Huiyun Zhang
- Beijing Chao-Yang Hospital, Dept. of Oncology, Capital Medical University, Beijing, China
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian Univ. of Traditional Chinese Medicine, Fuzhou, China.,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fuzhou, China
| | - Jiannan Yao
- Beijing Chao-Yang Hospital, Dept. of Oncology, Capital Medical University, Beijing, China
| |
Collapse
|
126
|
Remash D, Prince DS, McKenzie C, Strasser SI, Kao S, Liu K. Immune checkpoint inhibitor-related hepatotoxicity: A review. World J Gastroenterol 2021; 27:5376-5391. [PMID: 34539139 PMCID: PMC8409159 DOI: 10.3748/wjg.v27.i32.5376] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/28/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
The application of immune checkpoint inhibitors (ICI) in advanced cancer has been a major development in the last decade. The indications for ICIs are constantly expanding into new territory across different cancers, disease stages and lines of therapy. With this increased use, adverse events including immune checkpoint inhibitor-related hepatotoxicity (ICH) have emerged as an important clinical problem. This along with the introduction of ICI as first- and second-line treatments for advanced hepatocellular carcinoma makes ICH very relevant to gastroenterologists and hepatologists. The incidence of ICH varies between 1%-20% depending on the number, type and dose of ICI received. Investigation and management generally involve excluding differential diagnoses and following a stepwise escalation of withholding or ceasing ICI, corticosteroid treatment and adding other immunosuppressive agents depending on the severity of toxicity. The majority of patients with ICH recover and some may even safely recommence ICI therapy. Guideline recommendations are largely based on evidence derived from retrospective case series which highlights a priority for future research.
Collapse
Affiliation(s)
- Devika Remash
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
| | - David S Prince
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
| | - Catriona McKenzie
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- New South Wales Health Pathology, New South Wales Health, Sydney 2050, NSW, Australia
| | - Simone I Strasser
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
| | - Steven Kao
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
- Medical Oncology, Chris O’Brien Lifehouse, Sydney 2050, NSW, Australia
| | - Ken Liu
- AW Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Sydney 2050, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney 2006, NSW, Australia
| |
Collapse
|
127
|
Giustozzi M, Becattini C, Roila F, Agnelli G, Mandalà M. Vascular events with immune checkpoint inhibitors in melanoma or non-small cell lung cancer: A systematic review and meta-analysis. Cancer Treat Rev 2021; 100:102280. [PMID: 34438237 DOI: 10.1016/j.ctrv.2021.102280] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/17/2022]
Abstract
The incidence of venous and arterial thromboembolic events in advanced cancer patients treated with immune checkpoint inhibitors (ICIs) has been sporadically reported. We performed a systematic review and meta-analysis to assess the rate of vascular events in patients with melanoma and non-small cell lung cancer (NSCLC) treated with ICIs. A systematic search of MEDLINE and EMBASE was performed to identify randomized clinical trials and prospective studies. The main outcomes were venous thromboembolism (VTE), stroke or systemic embolism (SE) and myocardial infarction (MI). Secondary outcomes were fatal VTE, fatal stroke or SE and fatal MI. Pooled proportions with 95% confidence intervals (CI) were calculated using random-effects models. A total of 59 trials, 25 in 5,578 patients with melanoma and 34 in 6,543 patients with NSCLC were included. In patients with melanoma, rates of VTE, stroke or SE and MI were 1.5% (95% CI 0.8-2.8), 1.7% (95% CI 0.8-3.7) and 0.4% (95% CI 0.2-0.9), respectively. In patients with NSCLC, corresponding rates were 1.9% (95% CI 1.2-3.2), 1.2% (95% CI 0.6-2.5), and 1.1% (95% CI 0.5-2.1), respectively. Rates of fatal VTE and MI were similar in melanoma and NSCLC patients. Rates of fatal stroke or SE were 1.9% (95% CI 0.4-9.5) and 0.7% (95% CI 0.2-2.3) in melanoma and NSCLC patients, respectively. Rates of VTE (3.1% vs. 1.1%) and myocardial infarction (3.4% Vs. 0.5%) were numerically higher in NSCLC patients treated with combined-ICIs vs mono-ICIs. Our study shows a not negligible rate of vascular events in patients with melanoma or NSCLC treated with ICIs.
Collapse
Affiliation(s)
- Michela Giustozzi
- Internal, Vascular and Emergency Medicine - Stroke Unit, University of Perugia, Perugia, Italy.
| | - Cecilia Becattini
- Internal, Vascular and Emergency Medicine - Stroke Unit, University of Perugia, Perugia, Italy
| | - Fausto Roila
- Unit of Medical Oncology, University of Perugia, Perugia, Italy
| | - Giancarlo Agnelli
- Internal, Vascular and Emergency Medicine - Stroke Unit, University of Perugia, Perugia, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, University of Perugia, Perugia, Italy
| |
Collapse
|
128
|
Piña Y, Evernden BR, Khushalani N, Margolin K, Tawbi H, Tran ND, Macaulay R, Forsyth P, Peguero E. Acute motor axonal neuropathy after ipilimumab and nivolumab treatment in melanoma brain metastases: A case report and review of the literature. SAGE Open Med Case Rep 2021; 9:2050313X211042215. [PMID: 34457306 PMCID: PMC8392782 DOI: 10.1177/2050313x211042215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022] Open
Abstract
The use of immune checkpoint inhibitors including ipilimumab and nivolumab has expanded for several tumors including melanoma brain metastasis. These have resulted in a growing spectrum of neurologic immune-related adverse events, including ones that are rare and difficult to diagnose and treat. Here, we present a patient with melanoma brain metastasis who was treated with immune checkpoint inhibitors and developed an Acute Motor Axonal Neuropathy. To our knowledge, this is the first case of Acute Motor Axonal Neuropathy as an immune-related adverse event associated with combination treatment of ipilimumab and nivolumab, who was successfully treated. A 28-year-old woman with metastatic BRAF V600E melanoma developed melanoma brain metastasis and was enrolled on Checkmate 204, a Phase 2 clinical trial using ipilimumab (3 mg/kg intravenous) and nivolumab (1 mg/kg intravenous) every 3 weeks for four cycles, followed by monotherapy with nivolumab (240 mg intravenous) every 2 weeks. A few days after Cycle 2 of ipilimumab and nivolumab, she developed a pure motor axonal neuropathy consistent with Acute Motor Axonal Neuropathy. She was treated with several immunosuppressive treatments including high dose methylprednisolone, immune globulin, and infliximab, and her motor neuropathy eventually improved several months after onset of symptoms. Unfortunately, she had progression of her systemic disease and died several months later. This is the first case reported of Acute Motor Axonal Neuropathy associated with ipilimumab and nivolumab, successfully treated with immune-suppressive therapy. As the field of immunotherapy expands with the increasing use of the immune checkpoint inhibitors, it is critical to increase our knowledge and understanding of the neurologic immune-related adverse events associated with immune checkpoint inhibitors. This includes the spectrum of rare neurologic immune-related adverse events, which can be quite difficult to recognize and treat. Early consultations with neurology may expedite a diagnosis and treatment plan in patients with unexplained weakness receiving immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Yolanda Piña
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| | - Brittany R. Evernden
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| | - Nikhil Khushalani
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| | | | - Hussein Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nam D. Tran
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| | - Robert Macaulay
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| | - Peter Forsyth
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| | - Edwin Peguero
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
- H. Lee Moffitt Cancer Center (MCC) & Research Institute, Tampa, FL, USA
| |
Collapse
|
129
|
Tselikas L, de Baere T, Isoardo T, Susini S, Ser-Le Roux K, Polrot M, Adam J, Rouanne M, Zitvogel L, Moine L, Deschamps F, Marabelle A. Pickering emulsions with ethiodized oil and nanoparticles for slow release of intratumoral anti-CTLA4 immune checkpoint antibodies. J Immunother Cancer 2021; 8:jitc-2020-000579. [PMID: 32571995 PMCID: PMC7307549 DOI: 10.1136/jitc-2020-000579] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2020] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Intratumorous immunotherapy for cancer is currently thriving. The aim of such local strategy is to improve the therapeutic index of these treatments, for higher on-target/on-tumor activity and less on-target/off-tumor adverse events. Strategies allowing for slow release of anti-CTLA4 in the tumor microenvironment could improve their clinical efficacy.The purpose of the study was to develop a radiopaque delivery platform to improve the targeting and exposure of intratumorous anti-CTLA4 antibodies for cancer immunotherapy. METHODS Pickering emulsions of anti-CTLA4 antibodies were formulated with radiopaque ethiodized oil and poly-lactic-co-glycolic acid (PLGA) nanoparticles. We characterized the microscopic aspect and stability of such emulsions using Turbiscan. We monitored the release of anti-CTLA4 over time from these emulsions and evaluated their structure using mass spectrometry. We then tested the functionality of the released antibodies by preforming ex vivo competitive binding assays. Finally, we assessed the in vivo efficacy of intratumorous anti-CTLA4 Pickering emulsions. RESULTS Pickering emulsions of ethiodized oil and PLGA nanoparticles (PEEPs) resulted in a radiopaque water-in-oil emulsion with average internal phase droplet size of 42±5 µm at day 7. Confocal microscopy showed that anti-CTLA4 antibodies were effectively encapsulated by ethiodized oil with PLGA nanoparticles located at the interface between the aqueous and the oily phase. Turbiscan analysis showed that emulsions were stable with continuous and progressive release of anti-CTLA4 antibodies reaching 70% at 3 weeks. Structural and functional analysis of the released antibodies did not show significant differences with native anti-CTLA4 antibodies. Finally, intratumorous anti-CTLA4 PEEPs were able to eradicate tumors and cure mice in a syngeneic immunocompetent preclinical tumor model. CONCLUSION Pickering emulsions of ethiodized oil and PLGA is an innovative radiopaque delivery platform that does not alter the functionality of anti-CTLA4 immune checkpoint antibodies. Beyond local anti-CTLA4 applications, these emulsions might be used with other therapeutic molecules for optimal intratumorous or intra-arterial delivery of novel cancer immunotherapies.
Collapse
Affiliation(s)
- Lambros Tselikas
- Interventional Radiology, Gustave Roussy, Villejuif, France .,Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
| | | | - Thomas Isoardo
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
| | - Sandrine Susini
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
| | - Karine Ser-Le Roux
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/US23, Gustave Roussy, Villejuif, France
| | - Mélanie Polrot
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/US23, Gustave Roussy, Villejuif, France
| | - Julien Adam
- Pathology Department, Gustave Roussy, Villejuif, France
| | - Mathieu Rouanne
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France
| | | | - Laurence Moine
- Institut Galien, CNRS, Université Paris-Saclay, Châtenay-Malabry, France
| | | | - Aurélien Marabelle
- Laboratoire de Recherche Translationnelle en Immunothérapie (LRTI), INSERM U1015, Villejuif, France.,Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Drug Development Unit, Gustave Roussy, Villejuif, France
| |
Collapse
|
130
|
Weingarden AR, Rubin SJS, Gubatan J. Immune checkpoint inhibitor-mediated colitis in gastrointestinal malignancies and inflammatory bowel disease. World J Gastrointest Oncol 2021; 13:772-798. [PMID: 34457186 PMCID: PMC8371513 DOI: 10.4251/wjgo.v13.i8.772] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have markedly changed the landscape of cancer therapy. By re-invigorating the immune system against tumors, ICI provide novel therapeutic options for a broad variety of malignancies, including many gastrointestinal (GI) cancers. However, these therapies can also induce autoimmune-like side effects in healthy tissue across the body. One of the most common of these side effects is ICI-mediated colitis and diarrhea (IMC). Here, we review the incidence and risk of IMC in ICI therapy, with a focus on what is known regarding IMC in patients with GI malignancies. We also discuss data available on the use of ICI and risk of IMC in patients with pre-existing inflammatory bowel disease, as these patients may have increased risk of IMC due to their underlying intestinal pathology.
Collapse
Affiliation(s)
- Alexa R Weingarden
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Redwood City, CA 94063, United States
| | - Samuel J S Rubin
- Stanford University School of Medicine, Stanford University, Redwood City, CA 94063, United States
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Redwood City, CA 94063, United States
| |
Collapse
|
131
|
Cross-Validation of a Multiplex LC-MS/MS Method for Assaying mAbs Plasma Levels in Patients with Cancer: A GPCO-UNICANCER Study. Pharmaceuticals (Basel) 2021; 14:ph14080796. [PMID: 34451893 PMCID: PMC8401780 DOI: 10.3390/ph14080796] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Different liquid chromatography tandem mass spectrometry (LC-MS/MS) methods have been published for quantification of monoclonal antibodies (mAbs) in plasma but thus far none allowed the simultaneous quantification of several mAbs, including immune checkpoint inhibitors. We developed and validated an original multiplex LC-MS/MS method using a ready-to-use kit to simultaneously assay 7 mAbs (i.e., bevacizumab, cetuximab, ipilimumab, nivolumab, pembrolizumab, rituximab and trastuzumab) in plasma. This method was next cross-validated with respective reference methods (ELISA or LC-MS/MS). METHODS The mAbXmise kit was used for mAb extraction and full-length stable-isotope-labeled antibodies as internal standards. The LC-MS/MS method was fully validated following current EMA guidelines. Each cross validation between reference methods and ours included 16-28 plasma samples from cancer patients. RESULTS The method was linear from 2 to 100 µg/mL for all mAbs. Inter- and intra-assay precision was <14.6% and accuracy was 90.1-111.1%. The mean absolute bias of measured concentrations between multiplex and reference methods was 10.6% (range 3.0-19.9%). CONCLUSIONS We developed and cross-validated a simple, accurate and precise method that allows the assay of up to 7 mAbs. Furthermore, the present method is the first to offer a simultaneous quantification of three immune checkpoint inhibitors likely to be associated in patients.
Collapse
|
132
|
Mineiro dos Santos Garrett NF, Carvalho da Costa AC, Barros Ferreira E, Damiani G, Diniz dos Reis PE, Inocêncio Vasques C. Prevalence of dermatological toxicities in patients with melanoma undergoing immunotherapy: Systematic review and meta-analysis. PLoS One 2021; 16:e0255716. [PMID: 34358260 PMCID: PMC8345892 DOI: 10.1371/journal.pone.0255716] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
Background Checkpoint inhibitors have revolutionized advanced melanoma care; however, their cutaneous side effects have not been definitively elucidated. Objective To identify the prevalence of cutaneous toxicity in patients with melanoma treated with immune checkpoint inhibitors as monotherapy and/or in combination with chemotherapy and/or radiotherapy. Materials and methods We performed a systematic review and meta-analysis, which encompassed both clinical trials and observational studies describing the dermatological toxicities in patients treated with immune checkpoint inhibitors. The protocol was registered in the International Prospective Register of Systematic Review under the number CRD42018091915. The searches were performed using the CINAHL, Cochrane CENTRAL, LILACS, LIVIVO, PubMed, Scopus, and Web of Science databases. The methodological quality of the studies was evaluated with the JBI Critical Appraisal Checklist for Studies Reporting Prevalence Data Results A total of 9,802 articles were identified in the databases. The final sample comprised 39 studies. The evaluated drugs were ipilimumab, tremelimumab, pembrolizumab, and nivolumab. The results suggest that the most prevalent side effect was grade 1 and 2 pruritus (24%), followed by grade 1 and 2 rash (21%) and grade 1 and 2 vitiligo (10%). Conclusion The most prevalent side effects in patients treated with checkpoint inhibitors are pruritus, rash, and vitiligo, and they are rated mostly as grades 1 and 2 adverse events. Remarkably, vitiligo is most commonly found in patients treated with PD-1 inhibitors.
Collapse
Affiliation(s)
| | | | | | - Giovanni Damiani
- Clinical Dermatology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, United States of America
- Department of Drug Sciences, University of Padua, Padua, Italy
| | | | | |
Collapse
|
133
|
Identification of Immune Cell Infiltration in Murine Pheochromocytoma during Combined Mannan-BAM, TLR Ligand, and Anti-CD40 Antibody-Based Immunotherapy. Cancers (Basel) 2021; 13:cancers13163942. [PMID: 34439097 PMCID: PMC8393500 DOI: 10.3390/cancers13163942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/24/2021] [Accepted: 07/30/2021] [Indexed: 11/16/2022] Open
Abstract
Immunotherapy has become an essential component in cancer treatment. However, the majority of solid metastatic cancers, such as pheochromocytoma, are resistant to this approach. Therefore, understanding immune cell composition in primary and distant metastatic tumors is important for therapeutic intervention and diagnostics. Combined mannan-BAM, TLR ligand, and anti-CD40 antibody-based intratumoral immunotherapy (MBTA therapy) previously resulted in the complete eradication of murine subcutaneous pheochromocytoma and demonstrated a systemic antitumor immune response in a metastatic model. Here, we further evaluated this systemic effect using a bilateral pheochromocytoma model, performing MBTA therapy through injection into the primary tumor and using distant (non-injected) tumors to monitor size changes and detailed immune cell infiltration. MBTA therapy suppressed the growth of not only injected but also distal tumors and prolonged MBTA-treated mice survival. Our flow cytometry analysis showed that MBTA therapy led to increased recruitment of innate and adaptive immune cells in both tumors and the spleen. Moreover, adoptive CD4+ T cell transfer from successfully MBTA-treated mice (i.e., subcutaneous pheochromocytoma) demonstrates the importance of these cells in long-term immunological memory. In summary, this study unravels further details on the systemic effect of MBTA therapy and its use for tumor and metastasis reduction or even elimination.
Collapse
|
134
|
Navani V, Graves MC, Marchett GC, Mandaliya H, Bowden NA, van der Westhuizen A. Overall survival in metastatic melanoma correlates with pembrolizumab exposure and T cell exhaustion markers. Pharmacol Res Perspect 2021; 9:e00808. [PMID: 34129290 PMCID: PMC8204864 DOI: 10.1002/prp2.808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
Trial data support an absence of an exposure-survival relationship for pembrolizumab. As these relationships remain unexamined in a real-world setting, we determined them in metastatic melanoma prospectively in an observational study. Translational objectives included identifying biomarkers of progressive disease (PD). Checkpoint blockade naïve patients receiving 2 mg/kg Q3W pembrolizumab had pharmacokinetic and clinical outcome data collected. Trough, a valid surrogate for drug exposure, was assessed using ELISA. T-cell exhaustion and chemokine markers were determined using flow cytometry. Geometric means of exposures and biomarkers were tested against objective response groups using one-way ANOVA. The cohort was split by the median into high versus low pembrolizumab exposure groups. Kaplan-Meier progression-free survival (PFS) and overall survival (OS) curves were estimated for high versus low exposure, compared using the log rank test. The high pembrolizumab exposure group (n = 14) experienced substantially longer median OS (not reached vs. 48 months, p = .014), than the low exposure group (n = 14). A similar positive exposure PFS relationship was found (median not reached vs. 48 months, p = .045). The frequency of TIM-3 expression on CD4+ T cells was significantly higher in PD (mean 27.8%) than complete response (CR) (13.38%, p = .01) and partial response (12.4%, p = .05). There was a near doubling of CXCR6 and TIM-3 co-expression on CD4+ T cells in PD (mean 23.3%) versus CR (mean 11.4, p = .003) and partial response (9.8%, p = .0001). We describe positive exposure-PFS and exposure-OS relationships for pembrolizumab in metastatic melanoma. TIM-3, alongside co-expression of CXCR6 and TIM-3 on circulating CD4+ T cells are potential bio markers of treatment failure.
Collapse
Affiliation(s)
- Vishal Navani
- Calvary Mater Hospital NewcastleWaratahNSWAustralia
- Centre for Drug Repurposing and Medicines ResearchUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNSWAustralia
| | - Moira C. Graves
- Centre for Drug Repurposing and Medicines ResearchUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNSWAustralia
| | | | | | - Nikola A. Bowden
- Centre for Drug Repurposing and Medicines ResearchUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNSWAustralia
| | - Andre van der Westhuizen
- Calvary Mater Hospital NewcastleWaratahNSWAustralia
- Centre for Drug Repurposing and Medicines ResearchUniversity of Newcastle and Hunter Medical Research InstituteCallaghanNSWAustralia
| |
Collapse
|
135
|
Abstract
Hepatocellular carcinoma (HCC) is a prevalent disease with a progression that is modulated by the immune system. Systemic therapy is used in the advanced stage and until 2017 consisted only of antiangiogenic tyrosine kinase inhibitors (TKIs). Immunotherapy with checkpoint inhibitors has shown strong anti-tumour activity in a subset of patients and the combination of the anti-PDL1 antibody atezolizumab and the VEGF-neutralizing antibody bevacizumab has or will soon become the standard of care as a first-line therapy for HCC, whereas the anti-PD1 agents nivolumab and pembrolizumab are used after TKIs in several regions. Other immune strategies such as adoptive T-cell transfer, vaccination or virotherapy have not yet demonstrated consistent clinical activity. Major unmet challenges in HCC checkpoint immunotherapy are the discovery and validation of predictive biomarkers, advancing treatment to earlier stages of the disease, applying the treatment to patients with liver dysfunction and the discovery of more effective combinatorial or sequential approaches. Combinations with other systemic or local treatments are perceived as the most promising opportunities in HCC and some are already under evaluation in large-scale clinical trials. This Review provides up-to-date information on the best use of currently available immunotherapies in HCC and the therapeutic strategies under development.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver Unit and HPB Oncology Area, Clinica Universidad de Navarra-IDISNA and CIBEREHD, Pamplona, Spain.
| | - Pablo Sarobe
- Program of Immunology and Immunotherapy, CIMA de la Universidad de Navarra, IDISNA and CIBEREHD, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Program of Immunology and Immunotherapy, CIMA de la Universidad de Navarra, IDISNA and CIBEREHD, Pamplona, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, CIMA de la Universidad de Navarra, IDISNA and CIBEREHD, Pamplona, Spain
- Department of Immunology and Immunotherapy, Clinica Universidad de Navarra-IDISNA and CIBERONC, Pamplona, Spain
| |
Collapse
|
136
|
Sullivan RJ, Weber JS. Immune-related toxicities of checkpoint inhibitors: mechanisms and mitigation strategies. Nat Rev Drug Discov 2021; 21:495-508. [PMID: 34316029 DOI: 10.1038/s41573-021-00259-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
The immune-related adverse events associated with treatment with immune checkpoint inhibitors result in significant morbidity for patients as well as considerable cost to the health-care system, and can limit the use of these beneficial drugs. Understanding the mechanisms of these side effects and how they can be separated from the antitumour effects of immune checkpoint inhibitors, as well as identifying biomarkers that predict the development of immune-related toxicities, will facilitate the conduct of trials to limit their onset and improve patient outcomes. In this Review, we discuss the different types of immune-related adverse events and how their treatment and identification of possible predictive biomarkers may shed light on their mechanisms, and describe possible strategies and targets for prophylactic and therapeutic intervention to mitigate them.
Collapse
Affiliation(s)
- Ryan J Sullivan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey S Weber
- Laura and Isaac Perlmutter Comprehensive Cancer Center, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
137
|
Impact of open-label versus blinded study design on patient-reported outcomes data in randomized clinical trials of immunotherapy in advanced or metastatic cancer patients: a systematic review. Qual Life Res 2021; 31:645-657. [PMID: 34283382 DOI: 10.1007/s11136-021-02945-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
AIM A systematic literature review of immuno-oncology trials was conducted to assess the potential impact of open-label vs double-blind trial design on patient-reported outcome (PRO) data. METHODS A systematic search of indexed literature published from January 2009 to May 2019 was conducted using PubMed/MEDLINE, Cochrane Library, and EMBASE database. All randomized clinical trials (RCTs) of immuno-oncology therapies on advanced cancer patients reporting PRO data were identified. Descriptive analyses were performed to quantify differences at baseline and over time, by the type of study, regarding questionnaire completion rate and PRO scores. RESULTS In total, 23 studies were retained (15 open-label, 8 blinded). At baseline, no difference in completion rate was observed between arms irrespective of trial design (absolute mean difference of 2.8% and 2.2% for open label and blinded studies, respectively). No clinically significant difference in baseline PRO scores was observed between arms. Over time, impact on PRO scores could not be identified due to the limited number of studies, heterogeneity of questionnaires and tumor types. CONCLUSIONS Trial design had no impact on PRO completion rate or baseline scores. Future research should involve analyses by specific cancer types and ideally compare individual data from two similar RCTs (blinded vs. open-label).
Collapse
|
138
|
Crispo A, Corradin MT, Giulioni E, Vecchiato A, Del Fiore P, Queirolo P, Spagnolo F, Vanella V, Caracò C, Tosti G, Pennacchioli E, Giudice G, Nacchiero E, Quaglino P, Ribero S, Giordano M, Marussi D, Barruscotti S, Guida M, De Giorgi V, Occelli M, Grosso F, Cairo G, Gatti A, Massa D, Atzori L, Calvani N, Fabrizio T, Mastrangelo G, Toffolutti F, Celentano E, Budroni M, Gandini S, Rossi CR, Testori A, Palmieri G, Ascierto PA. Real Life Clinical Management and Survival in Advanced Cutaneous Melanoma: The Italian Clinical National Melanoma Registry Experience. Front Oncol 2021; 11:672797. [PMID: 34307142 PMCID: PMC8298066 DOI: 10.3389/fonc.2021.672797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Background Cutaneous melanoma (CM) is one of the most aggressive types of skin cancer. Currently, innovative approaches such as target therapies and immunotherapies have been introduced in clinical practice. Data of clinical trials and real life studies that evaluate the outcomes of these therapeutic associations are necessary to establish their clinical utility. The aim of this study is to investigate the types of oncological treatments employed in the real-life clinical management of patients with advanced CM in several Italian centers, which are part of the Clinical National Melanoma Registry (CNMR). Methods Melanoma-specific survival and overall survival were calculated. Multivariate Cox regression models were used to estimate the hazard ratios adjusting for confounders and other prognostic factors. Results The median follow-up time was 36 months (range 1.2-185.1). 787 CM were included in the analysis with completed information about therapies. All types of immunotherapy showed a significant improved survival compared with all other therapies (p=0.001). 75% was the highest reduction of death reached by anti-PD-1 (HR=0.25), globally immunotherapy was significantly associated with improved survival, either for anti-CTLA4 monotherapy or combined with anti-PD-1 (HR=0.47 and 0.26, respectively) and BRAFI+MEKI (HR=0.62). Conclusions The nivolumab/pembrolizumab in combination of ipilimumab and the addition of ant-MEK to the BRAFi can be considered the best therapies to improve survival in a real-world-population. The CNMR can complement clinical registries with the intent of improving cancer management and standardizing cancer treatment.
Collapse
Affiliation(s)
- Anna Crispo
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | | | - Erika Giulioni
- Dermatology Department, Azienda Sanitaria Friuli Occidentale, Pordenone, Italy
| | | | | | - Paola Queirolo
- IRCCS Ospedale Policlinico San Martino, Genova, Italy.,Istituto Europeo di Oncologia - IRCCS, Milano, Italy
| | | | - Vito Vanella
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Corrado Caracò
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Giulio Tosti
- Istituto Europeo di Oncologia - IRCCS, Milano, Italy
| | | | - Giuseppe Giudice
- Plastic and Reconstructive Surgery Department, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Eleonora Nacchiero
- Plastic and Reconstructive Surgery Department, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Pietro Quaglino
- Clinica Dermatologica, Dipartimento di Scienze Mediche, Università di Torino, Torino, Italy
| | - Simone Ribero
- Clinica Dermatologica, Dipartimento di Scienze Mediche, Università di Torino, Torino, Italy
| | - Monica Giordano
- Oncology Department, Ospedale Sant'Anna di Como, Como, Italy
| | - Desire Marussi
- Oncology Department, Ospedale Sant'Anna di Como, Como, Italy
| | | | - Michele Guida
- IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | | | - Marcella Occelli
- Oncology Department, Azienda ospedaliera Santa Croce e Carle, Cuneo, Italy
| | - Federica Grosso
- Mesothelioma Unit, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Giuseppe Cairo
- Oncology Department, ospedale "Vito Fazzi" di Lecce, Lecce, Italy
| | - Alessandro Gatti
- ULSS 2 Marca Trevigiana Ospedale Ca' Foncello Treviso, Treviso, Italy
| | - Daniela Massa
- Gruppo melanoma e tumori rari, Oncology Department, PO A Businco ARNAS G. Brotzu, Cagliari, Italy
| | - Laura Atzori
- Dermatology Clinic, Department Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Nicola Calvani
- Oncology Department, Presidio Ospedaliero "Senatore Antonio Perrino", Brindisi, Italy
| | - Tommaso Fabrizio
- IRCCS Centro di Riferimento Oncologico Basilicata, Rionero in Vulture, Italy
| | | | | | - Egidio Celentano
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Mario Budroni
- Registro Tumori Provincia di Sassari, Azienda Ospedaliera Universitaria Sassari, Sassari, Italy
| | - Sara Gandini
- Istituto Europeo di Oncologia - IRCCS, Milano, Italy
| | - Carlo Riccardo Rossi
- Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy.,Dermatology Clinic, Università degli studi di Padova, Padova, Italy
| | | | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | | |
Collapse
|
139
|
Dimitriou F, Long G, Menzies A. Novel adjuvant options for cutaneous melanoma. Ann Oncol 2021; 32:854-865. [DOI: 10.1016/j.annonc.2021.03.198] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/13/2021] [Accepted: 03/09/2021] [Indexed: 01/10/2023] Open
|
140
|
Dalle S, Mortier L, Corrie P, Lotem M, Board R, Arance AM, Meiss F, Terheyden P, Gutzmer R, Buysse B, Oh K, Brokaw J, Le TK, Mathias SD, Scotto J, Lord-Bessen J, Moshyk A, Kotapati S, Middleton MR. Long-term real-world experience with ipilimumab and non-ipilimumab therapies in advanced melanoma: the IMAGE study. BMC Cancer 2021; 21:642. [PMID: 34051732 PMCID: PMC8164785 DOI: 10.1186/s12885-021-08032-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/15/2021] [Indexed: 12/14/2022] Open
Abstract
Background Ipilimumab has shown long-term overall survival (OS) in patients with advanced melanoma in clinical trials, but robust real-world evidence is lacking. We present long-term outcomes from the IMAGE study (NCT01511913) in patients receiving ipilimumab and/or non-ipilimumab (any approved treatment other than ipilimumab) systemic therapies. Methods IMAGE was a multinational, prospective, observational study assessing adult patients with advanced melanoma treated with ipilimumab or non-ipilimumab systemic therapies between June 2012 and March 2015 with ≥3 years of follow-up. Adjusted OS curves based on multivariate Cox regression models included covariate effects. Safety and patient-reported outcomes were assessed. Results Among 1356 patients, 1094 (81%) received ipilimumab and 262 (19%) received non-ipilimumab index therapy (systemic therapy [chemotherapy, anti–programmed death 1 antibodies, or BRAF ± MEK inhibitors], radiotherapy, and radiosurgery). In the overall population, median age was 64 years, 60% were male, 78% were from Europe, and 78% had received previous treatment for advanced melanoma. In the ipilimumab-treated cohort, 780 (71%) patients did not receive subsequent therapy (IPI-noOther) and 314 (29%) received subsequent non-ipilimumab therapy (IPI-Other) on study. In the non-ipilimumab–treated cohort, 205 (78%) patients remained on or received other subsequent non-ipilimumab therapy (Other-Other) and 57 (22%) received subsequent ipilimumab therapy (Other-IPI) on study. Among 1151 patients who received ipilimumab at any time during the study (IPI-noOther, IPI-Other, and Other-IPI), 296 (26%) reported CTCAE grade ≥ 3 treatment-related adverse events, most occurring in year 1. Ipilimumab-treated and non-ipilimumab–treated patients who switched therapy (IPI-Other and Other-IPI) had longer OS than those who did not switch (IPI-noOther and Other-Other). Patients with prior therapy who did not switch therapy (IPI-noOther and Other-Other) showed similar OS. In treatment-naive patients, those in the IPI-noOther group tended to have longer OS than those in the Other-Other group. Patient-reported outcomes were similar between treatment cohorts. Conclusions With long-term follow-up (≥ 3 years), safety and OS in this real-world population of patients treated with ipilimumab 3 mg/kg were consistent with those reported in clinical trials. Patient-reported quality of life was maintained over the study period. OS analysis across both pretreated and treatment-naive patients suggested a beneficial role of ipilimumab early in treatment. Trial registration ClinicalTrials.gov, NCT01511913. Registered January 19, 2012 – Retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT01511913 Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08032-y.
Collapse
Affiliation(s)
- Stéphane Dalle
- Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, 69495, Pierre-Bénite, France.
| | - Laurent Mortier
- Université de Lille, INSERM U1189, CHRU Lille, 59037, Lille, France
| | - Pippa Corrie
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, CB0 2QQ, UK
| | - Michal Lotem
- Hadassah Hebrew University Hospital, 91120, Jerusalem, Israel
| | - Ruth Board
- Royal Preston Hospital, Preston, PR2 9HT, UK
| | | | - Frank Meiss
- Department of Dermatology, Faculty of Medicine, Medical Center - University of Freiburg, 79104, Freiburg, Germany
| | | | - Ralf Gutzmer
- Medizinische Hochschule Hannover, 30625, Hanover, Germany
| | | | - Kelly Oh
- Syneos Health, Morrisville, NC, 27560, USA
| | - Jane Brokaw
- Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | - T Kim Le
- Bristol Myers Squibb, Princeton, NJ, 08543, USA
| | | | | | | | | | | | | |
Collapse
|
141
|
Phase Ib/II trial testing combined radiofrequency ablation and ipilimumab in uveal melanoma (SECIRA-UM). Melanoma Res 2021; 30:252-260. [PMID: 31895753 DOI: 10.1097/cmr.0000000000000653] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Approximately, 50% of patients with uveal melanoma develop distant metastasis for which no standard therapy is established. In contrast to cutaneous melanoma, the anti-CTLA-4 antibody ipilimumab showed no clinical activity in uveal melanoma. Liver directed therapies improve local control, but fail to show overall survival (OS) benefit. Preclinical experiments demonstrated that radiofrequency ablation (RFA) induced durable responses in combination with anti-CTLA-4. The aim of this phase Ib/II study was to assess safety and efficacy of RFA plus ipilimumab in uveal melanoma. Patients underwent RFA of one liver lesion and subsequently received four courses ipilimumab 0.3, 3 or 10 mg/kg every 3 weeks in a 3 + 3 design. Primary endpoints were safety in terms of dose limiting toxicities per cohort to define the recommended phase II dose (RP2D) in the phase Ib part and confirmed the objective response rate and disease control rate (DCR) of non-RFA lesions in the phase II part. Secondary endpoints were progression-free survival (PFS) and OS. Ipilimumab 10 mg/kg + RFA was initially defined as the RP2D. However, after 19 patients, the study was amended to adjust the RP2D to ipilimumab 3 mg/kg + RFA, because 47% of patients treated with 10 mg/kg had developed grade 3 colitis. In the 3 mg/kg cohort, also 19 patients have been treated. Immunotherapy-related grade ≥3 adverse events were observed in 53% of patients in the 10 mg/kg cohort versus 32% in the 3 mg/kg cohort. No confirmed objective responses were observed; the confirmed DCR was 5% in the 10 mg/kg cohort and 11% in the 3 mg/kg cohort. Median PFS was 3 months and comparable for both cohorts, median OS was 14.2 months for the 10 mg/kg cohort versus 9.7 months for the 3 mg/kg cohort. Combining RFA with ipilimumab 3 mg/kg was well tolerated, but showed very limited clinical activity in uveal melanoma.
Collapse
|
142
|
Hu J, Tian R, Ma Y, Zhen H, Ma X, Su Q, Cao B. Risk of Cardiac Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Regimens: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:645245. [PMID: 34123795 PMCID: PMC8190385 DOI: 10.3389/fonc.2021.645245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Background We performed a systematic review and meta-analysis to evaluate the risks of cardiac adverse events in solid tumor patients treated with monotherapy of immune checkpoint inhibitors (ICIs) or combined therapy of ICIs plus chemotherapy. Methods Eligible studies were selected through the following databases: PubMed, Embase and clinical trials (https://clinicaltrials.gov.) and included phase III/IV randomized controlled trials (RCTs) involving patients with the solid tumor treated with ICIs. The data was analyzed by using Review Manager (version5.3), Stata (version 15.1). Results Among 2,551 studies, 25 clinical trials including 20,244 patients were qualified for the meta-analysis. Compared with PD-1 inhibitor (nivolumab) or CTLA-4 inhibitor (ipilimumab), PD-1 inhibitor (nivolumab) plus CTLA-4 inhibitor (ipilimumab) combined therapy showed significant increase in grade 5 arrhythmology (OR 3.90, 95% CI: 1.08–14.06, p = 0.603). PD-1 inhibitor plus chemotherapy show significant increase in grades 1–5 myocardial disease (OR 5.09, 95% CI: 1.11–23.32, p = 1.000). Compared with chemotherapy, PD-1 inhibitor (nivolumab) or CTLA-4 inhibitor (ipilimumab), PD-1 inhibitor (nivolumab) plus CTLA-4 inhibitor (ipilimumab) combined therapy show significant increase in grades 1–5 arrhythmology (OR 2.49, 95% CI: 1.30–4.78, p = 0.289). Conclusions Our meta-analysis demonstrated that PD-1 inhibitor plus CTLA-4 inhibitor can result in a higher risk of grade 5 arrhythmology in comparison with PD-1/CTLA-4 inhibitor alone, and a higher risk of grade 5 arrhythmology in comparison with chemotherapy. PD-1 inhibitor plus chemotherapy treatment could increase the risk of all-grade myocardial disease compared with chemotherapy. However, in most cases, there was no significant increase of risks of cardiovascular toxicity in PD-1/PD-L1 inhibitor monotherapy or PD-1/PD-L1 inhibitor plus chemotherapy compared with chemotherapy alone.
Collapse
Affiliation(s)
- Jiexuan Hu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ruyue Tian
- Department of Ultrasound, Aero Space Central Hospital, Beijing, China
| | - Yingjie Ma
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hongchao Zhen
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiao Ma
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qiang Su
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
143
|
Managing side effects of immune checkpoint inhibitors in breast cancer. Crit Rev Oncol Hematol 2021; 162:103354. [PMID: 34029683 DOI: 10.1016/j.critrevonc.2021.103354] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 02/06/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) represent a major development in cancer therapy. The indications for these agents continue to expand across malignancies and disease settings. For years breast cancer (BC) has been considered immunologically quiescent compared with other tumor types. However, recent findings highlighted the immunogenicity of some BCs and paved the way for clinical trials of immunotherapy in BC that led to recent landmark approvals. As a drawback, the safety profile of ICIs is shaped by a specific spectrum of immune-related adverse events (irAEs) that can vary according to ICI class and tumor histology. This review will discuss the epidemiology of these adverse events, their kinetics, risk factors and the most important aspects in their management. A particular focus will be put on BC as the current landscape of immunotherapy for this disease is rapidly increasing the number of people treated with ICIs, thus susceptible to irAEs.
Collapse
|
144
|
Ascierto PA, Del Vecchio M, Mackiewicz A, Robert C, Chiarion-Sileni V, Arance A, Lebbé C, Svane IM, McNeil C, Rutkowski P, Loquai C, Mortier L, Hamid O, Bastholt L, Dreno B, Schadendorf D, Garbe C, Nyakas M, Grob JJ, Thomas L, Liszkay G, Smylie M, Hoeller C, Ferraresi V, Grange F, Gutzmer R, Pikiel J, Hosein F, Simsek B, Maio M. Overall survival at 5 years of follow-up in a phase III trial comparing ipilimumab 10 mg/kg with 3 mg/kg in patients with advanced melanoma. J Immunother Cancer 2021; 8:jitc-2019-000391. [PMID: 32503946 PMCID: PMC7279645 DOI: 10.1136/jitc-2019-000391] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We have previously reported significantly longer overall survival (OS) with ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with advanced melanoma, with higher incidences of adverse events (AEs) at 10 mg/kg. This follow-up analysis reports a 5-year update of OS and safety. METHODS This randomized, multicenter, double-blind, phase III trial included patients with untreated or previously treated unresectable stage III or IV melanoma. Patients were randomly assigned (1:1) to ipilimumab 10 mg/kg or 3 mg/kg every 3 weeks for 4 doses. The primary end point was OS. RESULTS At a minimum follow-up of 61 months, median OS was 15.7 months (95% CI 11.6 to 17.8) at 10 mg/kg and 11.5 months (95% CI 9.9 to 13.3) at 3 mg/kg (HR 0.84, 95% CI 0.71 to 0.99; p=0.04). In a subgroup analysis, median OS of patients with asymptomatic brain metastasis was 7.0 months (95% CI 4.0 to 12.8) in the 10 mg/kg group and 5.7 months (95% CI 4.2 to 7.0) in the 3 mg/kg group. In patients with wild-type or mutant BRAF tumors, median OS was 13.8 months (95% CI 10.2 to 17.0) and 33.2 months (95% CI 19.4 to 45.2) in the 10 mg/kg group, and 11.2 months (95% CI 9.2 to 13.8) and 19.7 months (95% CI 11.6 to 25.3) in the 3 mg/kg group, respectively. The incidence of grade 3/4 treatment-related AEs was 36% in the 10 mg/kg group vs 20% in the 3 mg/kg group, and deaths due to treatment-related AEs occurred in four (1%) and two patients (1%), respectively. CONCLUSIONS This 61-month follow-up of a phase III trial showed sustained long-term survival in patients with advanced melanoma who started metastatic treatment with ipilimumab monotherapy, and confirmed the significant benefit for those who received ipilimumab 10 mg/kg vs 3 mg/kg. These results suggest the emergence of a plateau in the OS curve, consistent with previous ipilimumab studies. TRIAL REGISTRATION NUMBER NCT01515189.
Collapse
Affiliation(s)
- Paolo Antonio Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Michele Del Vecchio
- Unit of Melanoma Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Lombardia, Italy
| | - Andrzej Mackiewicz
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Center, Poznan Medical University, Poznan, Poland
| | - Caroline Robert
- Department of Medicine, Dermatology Service, Gustave Roussy, Villejuif and Paris-Sud-University, Le Kremlin-Bicêtre, France
| | | | - Ana Arance
- Hospital Clinic and Institut D'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Céleste Lebbé
- Université de Paris, INSERM, Dermatology and CIC, Saint Louis Hospital, Paris, France
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Herlev Hospital, Herlev, Denmark.,Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Catriona McNeil
- Chris O'Brien Lifehouse and Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie Institute-Oncology Center, Warsaw, Poland
| | - Carmen Loquai
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Laurent Mortier
- Clinique de Dermatologie, Unité d'Onco-Dermatologie, INSERM U1189, Centre Hospitalier Régional Universitaire de Lille, Hôpital Claude Huriez, Lille, France
| | - Omid Hamid
- Melanoma Center, The Angeles Clinic and Research Institute, Los Angeles, California, USA
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Brigitte Dreno
- Department of Oncodermatology, University Hospital Centre Nantes, Nantes, Pays de la Loire, France
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany.,Department of Dermatology, German Cancer Consortium, Heidelberg, Germany
| | - Claus Garbe
- Department of Dermatology, Eberhard Karls Universitat Tübingen, Tübingen, Baden-Württemberg, Germany
| | - Marta Nyakas
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Jean-Jacques Grob
- Dermatology and Skin Cancers Department, Aix-Marseille University, APHM, Marseille, France
| | - Luc Thomas
- Department of Dermatology, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Gabriella Liszkay
- Department of Oncodermatology, National Institute of Oncology, Budapest, Hungary
| | - Michael Smylie
- Department of Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Christoph Hoeller
- Division of General Dermatology and Dermato-Oncology, Medical University of Vienna, Vienna, Austria
| | - Virginia Ferraresi
- Unit of Medical Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Florent Grange
- Department of Dermatology, University Hospital Centre Reims, Reims, Champagne-Ardenne, France
| | - Ralf Gutzmer
- Operative Dermatology and Dermato-Oncology, Medizinische Hochschule Hannover, Hannover, Niedersachsen, Germany
| | - Joanna Pikiel
- Department of Oncology, Wojewodzkie Centrum Oncologii, Gdańsk, Poland
| | - Fareeda Hosein
- Oncology Clinical Development, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Burcin Simsek
- Oncology Clinical Development, Bristol-Myers Squibb Co, Princeton, New Jersey, USA.,Department of Biostatistics, Bristol-Myers Squibb Co, Princeton, New Jersey, USA
| | - Michele Maio
- Center for Immuno-Oncology, University Hospital of Siena, Instituto Toscano Tumori, Siena, Italy
| |
Collapse
|
145
|
Ananthakrishnan R, Green S, Previtali A, Liu R, Li D, LaValley M. Critical review of oncology clinical trial design under non-proportional hazards. Crit Rev Oncol Hematol 2021; 162:103350. [PMID: 33989767 DOI: 10.1016/j.critrevonc.2021.103350] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 05/03/2021] [Accepted: 05/08/2021] [Indexed: 12/16/2022] Open
Abstract
In trials of novel immuno-oncology drugs, the proportional hazards (PH) assumption often does not hold for the primary time-to-event (TTE) efficacy endpoint, likely due to the unique mechanism of action of these drugs. In practice, when it is anticipated that PH may not hold for the TTE endpoint with respect to treatment, the sample size is often still calculated under the PH assumption, and the hazard ratio (HR) from the Cox model is still reported as the primary measure of the treatment effect. Sensitivity analyses of the TTE data using methods that are suitable under non-proportional hazards (non-PH) are commonly pre-planned. In cases where a substantial deviation from the PH assumption is likely, we suggest designing the trial, calculating the sample size and analyzing the data, using a suitable method that accounts for non-PH, after gaining alignment with regulatory authorities. In this comprehensive review article, we describe methods to design a randomized oncology trial, calculate the sample size, analyze the trial data and obtain summary measures of the treatment effect in the presence of non-PH. For each method, we provide examples of its use from the recent oncology trials literature. We also summarize in the Appendix some methods to conduct sensitivity analyses for overall survival (OS) when patients in a randomized trial switch or cross-over to the other treatment arm after disease progression on the initial treatment arm, and obtain an adjusted or weighted HR for OS in the presence of cross-over. This is an example of the treatment itself changing at a specific point in time - this cross-over may lead to a non-PH pattern of diminishing treatment effect.
Collapse
Affiliation(s)
| | | | | | - Rong Liu
- Bristol-Myers Squibb (BMS), 300 Connell Drive, Berkeley Heights, NJ, 07922, United States
| | - Daniel Li
- BMS, Seattle, Washington, 98109, United States
| | - Michael LaValley
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, United States
| |
Collapse
|
146
|
Pires da Silva I, Ahmed T, Reijers ILM, Weppler AM, Betof Warner A, Patrinely JR, Serra-Bellver P, Allayous C, Mangana J, Nguyen K, Zimmer L, Trojaniello C, Stout D, Lyle M, Klein O, Gerard CL, Michielin O, Haydon A, Ascierto PA, Carlino MS, Lebbe C, Lorigan P, Johnson DB, Sandhu S, Lo SN, Blank CU, Menzies AM, Long GV. Ipilimumab alone or ipilimumab plus anti-PD-1 therapy in patients with metastatic melanoma resistant to anti-PD-(L)1 monotherapy: a multicentre, retrospective, cohort study. Lancet Oncol 2021; 22:836-847. [PMID: 33989557 DOI: 10.1016/s1470-2045(21)00097-8] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/09/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Anti-PD-1 therapy (hereafter referred to as anti-PD-1) induces long-term disease control in approximately 30% of patients with metastatic melanoma; however, two-thirds of patients are resistant and will require further treatment. We aimed to determine the efficacy and safety of ipilimumab plus anti-PD-1 (pembrolizumab or nivolumab) compared with ipilimumab monotherapy in patients who are resistant to anti-PD-(L)1 therapy (hereafter referred to as anti-PD-[L]1). METHODS This multicentre, retrospective, cohort study, was done at 15 melanoma centres in Australia, Europe, and the USA. We included adult patients (aged ≥18 years) with metastatic melanoma (unresectable stage III and IV), who were resistant to anti-PD-(L)1 (innate or acquired resistance) and who then received either ipilimumab monotherapy or ipilimumab plus anti-PD-1 (pembrolizumab or nivolumab), based on availability of therapies or clinical factors determined by the physician, or both. Tumour response was assessed as per standard of care (CT or PET-CT scans every 3 months). The study endpoints were objective response rate, progression-free survival, overall survival, and safety of ipilimumab compared with ipilimumab plus anti-PD-1. FINDINGS We included 355 patients with metastatic melanoma, resistant to anti-PD-(L)1 (nivolumab, pembrolizumab, or atezolizumab), who had been treated with ipilimumab monotherapy (n=162 [46%]) or ipilimumab plus anti-PD-1 (n=193 [54%]) between Feb 1, 2011, and Feb 6, 2020. At a median follow-up of 22·1 months (IQR 9·5-30·9), the objective response rate was higher with ipilimumab plus anti-PD-1 (60 [31%] of 193 patients) than with ipilimumab monotherapy (21 [13%] of 162 patients; p<0·0001). Overall survival was longer in the ipilimumab plus anti-PD-1 group (median overall survival 20·4 months [95% CI 12·7-34·8]) than with ipilimumab monotherapy (8·8 months [6·1-11·3]; hazard ratio [HR] 0·50, 95% CI 0·38-0·66; p<0·0001). Progression-free survival was also longer with ipilimumab plus anti-PD-1 (median 3·0 months [95% CI 2·6-3·6]) than with ipilimumab (2·6 months [2·4-2·9]; HR 0·69, 95% CI 0·55-0·87; p=0·0019). Similar proportions of patients reported grade 3-5 adverse events in both groups (59 [31%] of 193 patients in the ipilimumab plus anti-PD-1 group vs 54 [33%] of 162 patients in the ipilimumab group). The most common grade 3-5 adverse events were diarrhoea or colitis (23 [12%] of 193 patients in the ipilimumab plus anti-PD-1 group vs 33 [20%] of 162 patients in the ipilimumab group) and increased alanine aminotransferase or aspartate aminotransferase (24 [12%] vs 15 [9%]). One death occurred with ipilimumab 26 days after the last treatment: a colon perforation due to immune-related pancolitis. INTERPRETATION In patients who are resistant to anti-PD-(L)1, ipilimumab plus anti-PD-1 seemed to yield higher efficacy than ipilimumab with a higher objective response rate, longer progression-free, and longer overall survival, with a similar rate of grade 3-5 toxicity. Ipilimumab plus anti-PD-1 should be favoured over ipilimumab alone as a second-line immunotherapy for these patients with advanced melanoma. FUNDING None.
Collapse
Affiliation(s)
- Ines Pires da Silva
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Westmead Hospital, Sydney, NSW, Australia; Blacktown Hospital, Sydney, NSW, Australia
| | - Tasnia Ahmed
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | | | - Alison M Weppler
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | | | | | | | - Clara Allayous
- AP-HP Dermatology, INSERM U976, Université de Paris, Saint Louis Hospital, Paris, France
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Khang Nguyen
- Westmead Hospital, Sydney, NSW, Australia; Blacktown Hospital, Sydney, NSW, Australia
| | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Claudia Trojaniello
- Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | - Dan Stout
- Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Megan Lyle
- Cairns Hospital, James Cook University, Cairns, QLD, Australia
| | - Oliver Klein
- Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia
| | - Camille L Gerard
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Michielin
- Oncology Department, Lausanne University Hospital, Lausanne, Switzerland
| | - Andrew Haydon
- Alfred Hospital, Monash University, Melbourne, VIC, Australia
| | - Paolo A Ascierto
- Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Napoli, Italy
| | - Matteo S Carlino
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Westmead Hospital, Sydney, NSW, Australia; Blacktown Hospital, Sydney, NSW, Australia
| | - Celeste Lebbe
- AP-HP Dermatology, INSERM U976, Université de Paris, Saint Louis Hospital, Paris, France
| | - Paul Lorigan
- The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, University of Manchester, Manchester, UK
| | | | - Shahneen Sandhu
- Peter MacCallum Cancer Centre, University of Melbourne, Melbourne, VIC, Australia
| | - Serigne N Lo
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia
| | | | - Alexander M Menzies
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Royal North Shore Hospital, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia.
| |
Collapse
|
147
|
Tarhini AA, Kang N, Lee SJ, Hodi FS, Cohen GI, Hamid O, Hutchins LF, Sosman JA, Kluger HM, Eroglu Z, Koon HB, Lawrence DP, Kendra KL, Minor DR, Lee CB, Albertini MR, Flaherty LE, Petrella TM, Streicher H, Sondak VK, Kirkwood JM. Immune adverse events (irAEs) with adjuvant ipilimumab in melanoma, use of immunosuppressants and association with outcome: ECOG-ACRIN E1609 study analysis. J Immunother Cancer 2021; 9:jitc-2021-002535. [PMID: 33963015 PMCID: PMC8108687 DOI: 10.1136/jitc-2021-002535] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2021] [Indexed: 01/30/2023] Open
Abstract
Background The impact of immune-related adverse events (irAEs) occurring from adjuvant use of immunotherapy and of their management on relapse-free survival (RFS) and overall survival (OS) outcomes is currently not well understood. Patients and methods E1609 enrolled 1673 patients with resected high-risk melanoma and evaluated adjuvant ipilimumab 3 mg/kg (ipi3) and 10 mg/kg (ipi10) versus interferon-α. We investigated the association of irAEs and of use of immunosuppressants with RFS and OS for patients treated with ipilimumab (n=1034). Results Occurrence of grades 1–2 irAEs was associated with RFS (5 years: 52% (95% CI 47% to 56%) vs 41% (95% CI 31% to 50%) with no AE; p=0.006) and a trend toward improved OS (5 years: 75% (95% CI 71% to 79%) compared with 67% (95% CI 56% to 75%) with no AE; p=0.064). Among specific irAEs, grades 1–2 rash was most significantly associated with RFS (p=0.002) and OS (p=0.003). In multivariate models adjusting for prognostic factors, the most significant associations were seen for grades 1–2 rash with RFS (p<0.001, HR=0.70) and OS (p=0.01, HR=0.71) and for grades 1–2 endocrine+rash with RFS (p<0.001, HR=0.66) and OS (p=0.008, HR=0.7). Overall, grades 1–2 irAEs had the best prognosis in terms of RFS and OS and those with grades 3–4 had less RFS benefits and no OS advantage over no irAE. Patients experiencing grades 3–4 irAE had significantly higher exposure to corticosteroids and immunosuppressants than those with grades 1–2 (92% vs 60%; p<0.001), but no significant associations were found between corticosteroid and immunosuppressant use and RFS or OS. In investigating the impact of non-corticosteroid immunosuppressants, although there were trends toward better RFS and OS favoring cases who were not exposed, no significant associations were found. Conclusions Rash and endocrine irAEs were independent prognostic factors of RFS and OS in patients treated with adjuvant ipilimumab. Patients experiencing lower grade irAEs derived the most benefit, but we found no significant evidence supporting a negative impact of high dose corticosteroids and immunosuppressants more commonly used to manage grades 3–4 irAEs.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Center Inc, Tampa, Florida, USA
| | - Ni Kang
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Sandra J Lee
- Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - F Stephen Hodi
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Gary I Cohen
- Greater Baltimore Medical Center, Baltimore, Maryland, USA
| | - Omid Hamid
- The Angeles Clinic & Research Institute, A Cedars Sinai Affiliate, Los Angeles, California, USA
| | - Laura F Hutchins
- Department of Medicine, University of Arkansas for Medical Sciences (UAMS), Little Rock, Arkansas, USA
| | - Jeffrey A Sosman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
| | - Harriet M Kluger
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Zeynep Eroglu
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Center Inc, Tampa, Florida, USA
| | - Henry B Koon
- Case Western Reserve University, Cleveland, Ohio, USA
| | | | | | - David R Minor
- Sutter-California Pacific Medical Center, San Francisco, California, USA
| | - Carrie B Lee
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Lawrence E Flaherty
- Wayne State University and Karmanos Cancer Institute, Detroit, Michigan, USA
| | | | | | - Vernon K Sondak
- Departments of Cutaneous Oncology and Immunology, H. Lee Moffitt Cancer Center and Research Center Inc, Tampa, Florida, USA
| | | |
Collapse
|
148
|
Burton EM, Tawbi HA. Bispecific Antibodies to PD-1 and CTLA4: Doubling Down on T Cells to Decouple Efficacy from Toxicity. Cancer Discov 2021; 11:1008-1010. [PMID: 33947716 DOI: 10.1158/2159-8290.cd-21-0257] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Although combination anti-PD-1 and anti-CTLA4 mAbs have revolutionized outcomes for many cancers, their utility has been limited due to significant immune-related toxicities and the emergence of resistance. In this issue of Cancer Discovery, Dovedi and colleagues describe the development and preclinical testing of MEDI5752, a bispecific anti-PD-1/CTLA4 antibody designed to optimize therapeutic response by maximizing CTLA4 blockade on antigen-experienced T cells, thereby increasing efficacy and potentially minimizing toxicity.See related article by Dovedi et al., p. 1100.
Collapse
Affiliation(s)
- Elizabeth M Burton
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
149
|
Wilkes JG, Patel A, McClure E, Pina Y, Zager JS. Developments in therapy for brain metastases in melanoma patients. Expert Opin Pharmacother 2021; 22:1443-1453. [PMID: 33688795 DOI: 10.1080/14656566.2021.1900117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Introduction: Cutaneous melanoma brain metastases (MBM) are a major cause of morbidity and mortality. While cytotoxic agents, interferon, or interleukin-2, have been used with some success in extracranial disease, limited efficacy is demonstrated in MBM. The rare patient with long-term survival presented with limited intracranial disease amenable to surgery or radiation therapy. However, the development of targeted therapy and immunotherapy over the last decade has significantly improved overall survival in this formerly devastating presentation of metastatic melanoma.Areas covered: This article reviews the mechanism of brain metastasis, challenges with treating the central nervous system, historical treatment of MBM, and outcomes in clinical trials with targeted therapy and immunotherapy.Expert opinion: The MBM patient population now, more than ever, requires a multidisciplinary approach with surgery, radiation therapy, and the use of newer systemic therapies such as immunotherapy agents and targeted therapy agents. MBM has traditionally been excluded from clinical trials for systemic therapy due to poor survival. However, recent data show overall survival rates have significantly improved, supporting the need for inclusion of MBM patients in systemic therapy clinical trials. Understanding the mechanisms of therapeutic activity in the brain, resistance mechanisms, and the appropriate multi-modality treatment approach requires further investigation. Nevertheless, these therapies continue to give some hope to patients with historically poor survival.
Collapse
Affiliation(s)
- Justin G Wilkes
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA.,University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Ayushi Patel
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Erin McClure
- University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Yolanda Pina
- Department of Neuro-Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA.,University of South Florida Morsani College of Medicine, Tampa, FL, USA
| |
Collapse
|
150
|
Checkpoint Inhibitor-Induced Colitis-A Clinical Overview of Incidence, Prognostic Implications and Extension of Current Treatment Options. Pharmaceuticals (Basel) 2021; 14:ph14040367. [PMID: 33923423 PMCID: PMC8074139 DOI: 10.3390/ph14040367] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/19/2022] Open
Abstract
In recent years, anti-tumor immunotherapies have witnessed a major breakthrough with the emergence of immune checkpoint inhibitors (ICIs). However, the use of ICIs has also brought an era of a certain class of adverse events that differ from those of classical chemotherapies and are more reminiscent of autoimmune diseases. This article focuses exclusively on colitis as an irAE with emphasis on vulnerable patient groups, the prognostic significance of colitis, treatment, and new therapeutic approaches that may be applicable. Colitis itself is associated with a favorable oncological outcome of the underlying disease but is as well the most common irAE leading to discontinuation of therapy. Especially in vulnerable patient groups such as IBD patients and elderly patients, colitis occurs more frequently as a side effect. It is precisely in these two patient groups that side effects more often lead to discontinuation of therapy. Therefore, in addition to the current therapy of colitis through immunosuppression, the focus should also be on new forms of therapy of severe colitis, such as fecal transplantation or ileostomy creation.
Collapse
|