1
|
Sekar P, Ashok Kumar A, Hughes I, Mason R, Dzienis M. The safety of extended dosing pembrolizumab when compared to traditional dosing: A review and meta-analysis of retrospective studies. Lung Cancer 2025; 204:108585. [PMID: 40398109 DOI: 10.1016/j.lungcan.2025.108585] [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: 04/24/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND To minimise healthcare exposure in the COVID 19 era, 6-weekly extended dosing pembrolizumab became widely accepted as an alternative to 3-weekly dosing based on modelling studies. A review and meta-analysis were performed to assess the real-world safety of 6-weekly pembrolizumab in relation to 3-weekly dosing. METHODS Following PRISMA guidelines, we conducted a meta-analysis encompassing retrospective studies of solid organ malignancies comparing 6-weekly pembrolizumab with 3-weekly pembrolizumab including patients switched to 6-weekly from 3-weekly pembrolizumab ("switched"). Primary outcomes were relative risk estimates (RR) of grade 3-5 immune related adverse events (irAEs) for 6-weekly and "switched" pembrolizumab in relation to 3-weekly dosing. Secondary outcomes were pooled proportions of grade 3-5 irAEs for 6-weekly, 3-weekly and "switched" patients. A random effects logistic regression was used to estimate RR and the results were depicted in forest plots. RESULTS 10 studies were included with a total of 592 3-weekly, 520 6-weekly and 273 "switched" patients. 6-weekly pembrolizumab was not associated with increased rates of grade 3-5 irAEs when compared to 3-weekly dosing (RR 1.16; CI 0.85-1.58, p = 0.35). "Switched" 6-weekly patients had no significant difference in rates of grade 3-5 irAEs when compared to 3-weekly patients (RR 1.30; CI 0.87-2.07, p = 0.28). Total grade 3-5 irAEs for 6-weekly, 3-weekly and "switched" pembrolizumab patients were 15 %, 13 % and 16 %, respectively. CONCLUSION 6-weekly pembrolizumab is not associated with increased grade 3-5 irAEs compared to 3-weekly pembrolizumab. Switching patients to 6-weekly pembrolizumab from 3-weekly dosing was not associated with an increased risk of grade 3-5 irAEs.
Collapse
Affiliation(s)
- Prasanth Sekar
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, QLD 4215, Australia.
| | | | - Ian Hughes
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, QLD 4215, Australia
| | - Robert Mason
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, QLD 4215, Australia
| | - Marcin Dzienis
- Gold Coast University Hospital, 1 Hospital Blvd, Southport, QLD 4215, Australia
| |
Collapse
|
2
|
Price T, Lugowska I, Chawla SP, Falchook G, Subbiah V, Monzon JG, Arkenau HT, Hui M, Kuboki Y, Dziadziuszko R, Shibaki R, Hong MH, Tan D, Rocha Lima CM, Wang K, Hindoyan A, Shi W, Wong H, Kistler M, Prenen H. A phase I, open-label, multicentre, first-in-human study to evaluate safety, pharmacokinetics and efficacy of AMG 404, a PD-1 inhibitor, in patients with advanced solid tumours. BMJ Open 2025; 15:e088578. [PMID: 40316348 PMCID: PMC12049887 DOI: 10.1136/bmjopen-2024-088578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 04/04/2025] [Indexed: 05/04/2025] Open
Abstract
OBJECTIVE To evaluate the safety, tolerability, pharmacokinetics (PK) and preliminary antitumour activity of AMG 404, a fully human IgG1 monoclonal antibody targeting programmed cell death-1, in patients with advanced solid tumours. DESIGN First-in-human phase I study comprising eight dose expansion cohorts, including cohorts with microsatellite instability-high (MSI-H) tumours and non-small cell lung cancer with high programmed death-ligand 1 expression (NSCLC/PDL1-H, tumour proportion score ≥50%). SETTING Conducted across 28 global sites. PARTICIPANTS This study enrolled adult patients with histologically or cytologically confirmed metastatic or locally advanced solid tumours not amenable to curative treatment with surgery or radiation. The inclusion criteria included a life expectancy of >3 months, ≥1 measurable or evaluable lesion per modified Response Evaluation Criteria in Solid Tumours (RECIST) V.1.1, an Eastern Cooperative Oncology Group performance status of ≤2 and adequate haematological, renal and hepatic function. Patients with prior treatment with checkpoint inhibitors, primary brain tumour or untreated or symptomatic brain metastases and leptomeningeal disease and history of other malignancy within the past 2 years were excluded. INTERVENTIONS The planned doses were 240 mg, 480 mg and 1050 mg of AMG 404 administered every 4 weeks (Q4W). PRIMARY AND SECONDARY OUTCOME MEASURES Primary endpoints were dose-limiting toxicities (DLTs), treatment-emergent adverse events, treatment-related adverse events, changes in vital signs and clinical laboratory tests. Secondary endpoints included PK parameters, incidence of antidrug (AMG 404) antibodies and antitumour activity assessed per modified RECIST V.1.1 (objective response, duration of response, progression-free survival (PFS), disease control and duration of stable disease). RESULTS A total of 171 patients were enrolled; 168 were treated. Median (range) follow-up was 36.3 weeks (1.6-137.1). No DLTs were observed. Grade 3 and serious treatment-related adverse events occurred in 16 (9.5%) and 12 (7.1%) patients, respectively. The 480 mg Q4W dose was selected as the recommended phase II dose. AMG 404 serum exposure increased approximately dose proportionally. The objective response rate (80% CI) was 19.6% (15.7-24.1) for the overall population and 36.6% (26.4-47.8) and 30.8% (14.2-52.3) for cohorts with MSI-H tumours (n=41) and NSCLC/PDL1-H (n=13), respectively. The overall disease control rate (80% CI) was 54.8% (49.5-59.9). The median (80% CI) PFS was 3.7 (3.5-4.5) months for the overall population and 14.8 (9.0-not estimable) and 4.4 (2.2-9.7) months for cohorts with MSI-H tumours and NSCLC/PDL1-H, respectively. CONCLUSIONS AMG 404 monotherapy was tolerable at the tested doses, with encouraging antitumour activity observed across tumour types. TRIAL REGISTRATION NUMBER NCT03853109.
Collapse
MESH Headings
- Humans
- Female
- Male
- Middle Aged
- Aged
- Adult
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Neoplasms/drug therapy
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Lung Neoplasms/drug therapy
- Aged, 80 and over
- Immune Checkpoint Inhibitors/pharmacokinetics
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/therapeutic use
- Dose-Response Relationship, Drug
Collapse
Affiliation(s)
- Timothy Price
- The Queen Elizabeth Hospital, Woodville, Adelaide, Australia
| | - Iwona Lugowska
- Maria Sklodowska-Curie Institute of Oncology Warsaw, Warszawa, Poland
| | - Sant P Chawla
- Sarcoma Oncology Center, Santa Monica, California, USA
| | - Gerald Falchook
- Sarah Cannon Research Institute at HealthONE, Denver, Colorado, USA
| | - Vivek Subbiah
- University of Texas MD Anderson Cancer Centre, Houston, Texas, USA
| | | | | | - Mun Hui
- Chris O'Brien Lifehouse, Camperdown, New South Wales, Australia
| | | | | | | | - Min Hee Hong
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Daniel Tan
- National Cancer Centre Singapore, SingHealth, and Duke-NUS Medical School, Singapore
| | | | | | | | - Weibing Shi
- Amgen Inc San Francisco, South San Francisco, California, USA
| | - Hansen Wong
- Amgen Inc San Francisco, South San Francisco, California, USA
| | - Mira Kistler
- Amgen Inc San Francisco, South San Francisco, California, USA
| | - Hans Prenen
- University Hospital Antwerp, Edegem, Antwerp, Belgium
| |
Collapse
|
3
|
Guillen E, Barry S, Jost N, Ekman N, Knippel V, Kuhlmann-Gottke J, Maier J, Weise M, Laslop A, Anour R, van Zandbergen G, Kirsch-Stefan N. The Tailored Biosimilar Approach: Expectations and Requirements. Drugs 2025; 85:601-608. [PMID: 40169518 PMCID: PMC12031863 DOI: 10.1007/s40265-025-02168-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 04/03/2025]
Abstract
Regulatory approval of biosimilar medicines currently requires a combination of physicochemical and functional testing, pharmacokinetic data, and clinical efficacy studies (CES). In this article, we discuss the tailored biosimilar approach, which represents an evolution in regulatory thinking by moving away from the default requirement for CES in biosimilar approval. We explore how physicochemical and functional data can be predictive for clinical performance and address the limitations of CES for regulatory decision-making. We argue that, in most cases, the combination of a robust package of physicochemical and functional testing, with appropriately designed pharmacokinetic studies provides sufficient evidence to establish biosimilarity. Additionally, we provide our perspective on the requirements, expectations, and exceptions for future biosimilar approvals, outlining specific scenarios where additional clinical evidence may be necessary. These include cases where the mechanism of action is unknown or poorly characterized, when product heterogeneity cannot be adequately characterized, or where relevant safety or immunogenicity concerns arise with the reference product or biosimilar candidate. Finally, we aim to clarify the remaining concerns surrounding the tailored biosimilar approach, providing insights into the potential to streamline biosimilar development and regulatory approval.
Collapse
Affiliation(s)
- Elena Guillen
- Department of Clinical Pharmacology, Hospital Clínic, Barcelona, Spain
| | - Sean Barry
- Health Products Regulatory Authority, Dublin, Ireland
- Biologics Working Party, European Medicines Agency, Amsterdam, The Netherlands
| | - Nils Jost
- Division of Immunology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
- Biosimilar Medicinal Products Working Party, European Medicines Agency, Amsterdam, The Netherlands
| | - Niklas Ekman
- Finnish Medicines Agency, Helsinki, Finland
- Biologics Working Party, European Medicines Agency, Amsterdam, The Netherlands
- Biosimilar Medicinal Products Working Party, European Medicines Agency, Amsterdam, The Netherlands
| | - Verena Knippel
- Division of Immunology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Johanna Kuhlmann-Gottke
- Division of Immunology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Julia Maier
- Division of Immunology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Martina Weise
- Federal Institute for Drugs and Medical Devices, Bonn, Germany
- Biosimilar Medicinal Products Working Party, European Medicines Agency, Amsterdam, The Netherlands
| | - Andrea Laslop
- Austrian Medicines and Medical Devices Agency, Wien, Austria
- Scientific Advice Working Party, European Medicines Agency, Amsterdam, The Netherlands
| | - René Anour
- Austrian Medicines and Medical Devices Agency, Wien, Austria
- Biosimilar Medicinal Products Working Party, European Medicines Agency, Amsterdam, The Netherlands
| | - Ger van Zandbergen
- Division of Immunology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
- Institute for Immunology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nadine Kirsch-Stefan
- Centre for International Cooperation, WHO, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, 63225, Langen, Germany.
| |
Collapse
|
4
|
Tannock IF, de Vries EGE, Fojo A, Buyse M, Moja L. Dose optimisation to improve access to effective cancer medicines. Lancet Oncol 2025; 26:e171-e180. [PMID: 40049207 DOI: 10.1016/s1470-2045(24)00648-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/17/2024] [Accepted: 11/01/2024] [Indexed: 05/13/2025]
Abstract
Access to many cancer medicines on WHO's Essential Medicines List (EML) is restricted because of price, especially in low-income and middle-income countries (LMICs). Other cancer medicines that have been shown to improve survival, such as immune checkpoint inhibitors for lung cancer, are not included on the EML because approved doses and schedules exceed affordable prices in LMICs. Multiple strategies are therefore needed to reduce medicine costs or circumvent these problems, such as optimising doses and schedules. Cancer medicines are approved by regulatory agencies, such as the US Food and Drug Administration and the European Medicines Agency, following rigorous clinical trials. However, these approvals can involve dosing regimens and treatment schedules that, although effective in showing statistically significant benefits in trials, can be higher in intensity, frequency, or duration than is necessary to achieve meaningful improved survival. In clinical practice, these regimens can lead to concerns about balancing optimal therapeutic outcomes with the risk of side-effects, patient quality of life, and long-term health effects. Various types of evidence can, and should, be used to explore and show near-equivalence of beneficial outcomes from reduced-intensity treatments, including randomised clinical trials, dose-finding phase 1 and 2 studies, and pharmacokinetic and pharmacodynamic studies. The positive effects of proving that lower doses or less intensive schedules retain therapeutic activity include reduced toxicity and large price reductions, leading to better cost-effectiveness and greater access to treatments that improve survival. Beyond regulatory approvals, identification of regimens that have similar outcomes with reduced doses and less intense schedules should be a priority for clinicians and policy makers in the selection process to identify effective medicines at national and global levels.
Collapse
Affiliation(s)
- Ian F Tannock
- Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Elisabeth G E de Vries
- Department of Medical Oncology, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Antonio Fojo
- Division of Hematology/Oncology, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Marc Buyse
- International Drug Development Institute, Louvain-la-Neuve, Belgium
| | - Lorenzo Moja
- Health Products Policy and Standards, WHO, Geneva, Switzerland.
| |
Collapse
|
5
|
Kaushik M, Kapoor A, Singh H, Suresh P, Mulajkar D, Rathore A, Nair R, Nihanthy D, Mehrotra A, Patel A. Real world experience on patterns of usage and toxicity profile of immunotherapy drugs in Indian patients: A prospective observational study. Med J Armed Forces India 2025; 81:39-45. [PMID: 39872179 PMCID: PMC11762613 DOI: 10.1016/j.mjafi.2023.07.007] [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: 03/30/2023] [Accepted: 07/16/2023] [Indexed: 01/29/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) are now considered revolutionary agents in the treatment of various cancers. Prospective data are limited on the patterns of usage and toxicity profile of these drugs. We planned this study for addressing the same in Indian patients. Methods This prospective study was conducted over a period of 2 years. All patients who were treated with Nivolumab, pembrolizumab, atezolizumab, and durvalumab were included. Immune-related adverse events were recorded. Toxicities were graded and number of patients experiencing dose limiting toxicities was recorded. Results A total of 53 patients received one of the above four agents. Majority of patients were less than 60 years of age. Carcinoma lung was the most frequent malignancy followed by renal cell carcinoma, Hodgkin's Lymphoma, Urinary Bladder cancers, Malignant Melanoma, and Recurrent/Metastatic Head and neck cancer. Nivolumab was used in most of the study population followed by pembrolizumab. Majority of agents were used in second line. The frequency of all grade adverse events for fatigue, anemia, pneumonitis, skin rash, dyspnea, diarrhea, and hypothyroidism were (in %) 73.58, 62.26, 16.9, 11.32, 9.43, 9.43, and 7.55, respectively. No grade 5 toxicity was observed. None of the grade 3 or 4 toxicities led to treatment discontinuation. Statistically, no difference was found for all grade toxicities among ICI drugs and among the various lines of use. Conclusion Nivolumab was the commonest drug used in our cohort. Most of ICIs were used in second-line setting. Toxicities are in line with the published literature.
Collapse
Affiliation(s)
- M.R. Kaushik
- Classified Specialist (Medicine) & Medical Oncologist, INHS Asvini, Mumbai, India
| | - Amul Kapoor
- Consultant (Medicine & Medical Oncology), Army Hospital (R&R), New Delhi, India
| | - H.P. Singh
- Consultant (Medicine & Medical Oncology), Capitol Hospital, Jalandhar, India
| | - P. Suresh
- Senior Adviser (Medicine) & Medical Oncologist, Army Hospital (R&R), New Delhi, India
| | - Deepak Mulajkar
- Senior Adviser (Medicine) & Medical Oncologist, Army Hospital (R&R), New Delhi, India
| | - Anvesh Rathore
- Senior Adviser (Medicine) & Medical Oncologist, Army Hospital (R&R), New Delhi, India
| | - Rajesh Nair
- Classified Specialist (Medicine) & Medical Oncologist, Command Hospital (Central Command), Lucknow, India
| | - D.S. Nihanthy
- Senior Resident (Medical Oncology), Army Hospital (R&R), New Delhi, India
| | - Aarty Mehrotra
- Senior Resident (Medical Oncology), Army Hospital (R&R), New Delhi, India
| | - Amol Patel
- Senior Adviser (Medicine) & Medical Oncologist, INHS Asvini, Mumbai, India
| |
Collapse
|
6
|
Le Brun IC, Dalle S, Mortier L, Dereure O, Rat SD, Dutriaux C, Leccia MT, Legoupil D, Montaudié H, De Quatrebarbes J, Gaudy-Marqueste C, Maubec E, Saiag P, Pagès C, Possenti FB, Granel-Brocard F, Porcher R, Lefevre W, Lebbé C, Kempf E. Methods of nivolumab administration in advanced melanoma: A comparison of patients' clinical outcomes treated with flat dose or weight-adjusted dose, a multicenter observational study. Cancer 2025; 131:e35679. [PMID: 39645590 DOI: 10.1002/cncr.35679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Nivolumab obtained approval in advanced melanoma (AM) with weight-adjusted dose (WAD) administration (3 mg/kg/2 weeks). In 2018, the dosage regimen was changed to flat dose (FD) administration (240 mg/2 weeks or 480 mg/4 weeks) based on a modeling study, without clinical data. METHODS AM patients have been prospectively included in the French national multicenter MelBase database since 2013. First-line patients treated with nivolumab monotherapy were included in the WAD or FD groups of this study. The primary end point was the incidence of grade ≥3 immune-related adverse events (irAEs). Secondary end points were incidence of any grade irAEs, and overall survival (OS) and progression-free survival (PFS). Inverse probability of treatment weighting was used to balance groups on their baseline characteristics. RESULTS Between 2015 and 2022, 348 patients were included: 160 in the WAD and 188 in the FD groups. In the FD group, 45% and 27% of patients weighed <75 kg and >85 kg, respectively. Grade ≥3 and any grade irAEs rates were 13.1% versus 11.7% (p = .8) and 63.1% versus 67.0% (p = .5) in the WAD and FD groups, respectively. After weighting, median PFS was 3.1 and 3.7 months (hazard ratio [HR], 0.84; 95% confidence interval [CI], 0.65-1.07), and median OS was 24.8 and 37.0 months (HR, 0.74; 95% CI, 0.54-1.01) in the WAD and FD groups, respectively. CONCLUSIONS There was no difference in the incidence of severe irAEs and in median PFS between AM patients treated by WAD or FD nivolumab. The median OS between patient groups did not reach statistical significance.
Collapse
Affiliation(s)
- Iona Campo Le Brun
- Department of Medical Oncology, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Séphane Dalle
- INSERM 1052, Department of Dermatology, Hospices Civils de Lyon, Lyon-Sud hospital Pierre-Benite, Lyon University, Claude Bernard Lyon 1 University, CNRS 5286, Léon Bérard center, Cancer Research Center of Lyon, Lyon, France
| | - Laurent Mortier
- INSERM U1189, Department of Dermatology, Lille University Hospital, Université de Lille, Lille, France
| | - Olivier Dereure
- INSERM U1058, Department of Dermatology, University of Montpellier, Montpellier, France
| | - Sophie Dalac Rat
- Department of Dermatology, Dijon Bourgogne University Hospital, Dijon, France
| | | | - Marie-Thérèse Leccia
- Department of Dermatology, Allergology, Photobiology, Grenoble Alpes University Hospital, La Tronche, France
| | - Delphine Legoupil
- Department of Dermatology, Brest University Regional Hospital, Brest, France
| | - Henri Montaudié
- INSERM U1065, Department of Dermatology, Nice University Hospital, L'Archet Hospital, Nice, France
| | | | - Caroline Gaudy-Marqueste
- Dermatology and Skin Cancer Department, Aix-Marseille University, Assitance Publique des Hopitaux de Marseille, La Timone Hospital, Marseille, France
| | - Eve Maubec
- Department of Dermatology, Avicenne Hospital, Bobigny, France
| | - Philippe Saiag
- Department of General and Oncology Dermatology, Ambroise Paré Hospital, University Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Boulogne-Billancourt, France
| | - Cécile Pagès
- Department of Onco-Dermatology, University Cancer Institute-Oncopole, Toulouse, France
| | - Florence Brunet Possenti
- Department of Dermatology, Bichat-Claude-Bernard Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Florence Granel-Brocard
- Department of Dermatology, Nancy University Regional Hospital, Brabois Hospitals, Vandœuvre-lès-Nancy, France
| | - Raphaël Porcher
- Université Paris Cité and Université Sorbonne Paris Nord, Inserm, INRAE, Center for Research in Epidemiology and Statistics (CRESS), Centre d'Épidémiologie Clinique, Assistance Publique-Hôpitaux de Paris, Hôpital Hôtel Dieu, Paris, France
| | - Wendy Lefevre
- Saint Louis Hospital, Assistance Publique-Hôpitaux de Paris, MELBASE, Paris, France
| | - Célèste Lebbé
- INSERM U976, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Dermato-Oncology and CIC, Cancer Institute APHP nord Paris Cité, Saint Louis Hospital, Paris, France
| | - Emmanuelle Kempf
- Department of Medical Oncology, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France
| |
Collapse
|
7
|
Kumar A, Kapoor A, Noronha V, Patil VM, Joshi A, Menon N, Janu A, Mahajan A, Rajendra A, Agarawal A, Khaddar S, Rajpurohit A, Kashyap L, Srikant A, Panda G, Kota KK, Talreja V, Prabhash K. Efficacy and Safety of Low-Dose Nivolumab in Treatment of Advanced Solid Tumors: A Retrospective Audit from Resource-Constrained Settings. South Asian J Cancer 2025; 14:70-76. [PMID: 40124158 PMCID: PMC11925618 DOI: 10.1055/s-0044-1788649] [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: 08/01/2023] [Accepted: 07/02/2024] [Indexed: 03/25/2025] Open
Abstract
Background Immunotherapy has improved outcomes in many advanced solid tumors. In resource-constrained settings, less than 2% of patients can afford standard dose immunotherapy. A recent phase II study showed the efficacy of low-dose immunotherapy in this setting. We used low-dose immunotherapy on a compassionate basis in patients who had progressed on available standard treatment options and standard dose immunotherapy was not feasible. Patients and Methods We retrospectively collected data from the medical oncology department for consecutive patients who had initially received standard lines of therapy followed by low-dose immunotherapy (nivolumab 40 mg) on a compassionate basis. The demographic details, histology, prior treatment, clinical and radiological response, date of disease progression, date of death, and toxicity data were collected. Results A total of 54 consecutive patients, who received low-dose immunotherapy with nivolumab from January 1, 2018 to February 14, 2020, were included in this analysis; 4 patients were not radiologically evaluable. The median age was 50.4 years (range 35-74 years), male:female ratio was 6:1. The most common comorbidities were hypertension and diabetes seen in 12 (22.2%) and 6 (11.1%) patients, respectively. The majority of the patients (70.4%) were of head and neck cancer. The median follow-up was 4.5 months (range 0.5-11.7). Clinical benefit was observed in 18 (33.3%) patients. Partial response and stable disease were achieved in 9 (16.7%) and 5 (9.3%) patients, respectively. Median survival was not reached for these patients. Six months progression-free survival and overall survival were 100 versus 8.7% (hazard ratio [HR] 0.05, 95% confidence interval [CI]: 0.01-0.36; p = 0.003) and 100 versus 29.7% (HR 0.03, 95% CI: 0.00-0.95; p = 0.047), respectively, for responders and nonresponders. The side effects were manageable. Conclusion In resource-constrained settings, low-dose immunotherapy with nivolumab seems to be an effective treatment option. Further studies are warranted to evaluate this approach.
Collapse
Affiliation(s)
- Amit Kumar
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Akhil Kapoor
- Department of Medical Oncology, Mahamana Pandit Madan Mohan Malaviya Cancer Centre and Homi Bhabha Cancer Hospital, Tata Memorial Centre, Varanasi, Uttar Pradesh, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vijay M. Patil
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Janu
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Abhishek Mahajan
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Akhil Rajendra
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Amit Agarawal
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Satvik Khaddar
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Anu Rajpurohit
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Lakhan Kashyap
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Anne Srikant
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Gautam Panda
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kishore Kumar Kota
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vikas Talreja
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
8
|
Kulesza A, Couty C, Lemarre P, Thalhauser CJ, Cao Y. Advancing cancer drug development with mechanistic mathematical modeling: bridging the gap between theory and practice. J Pharmacokinet Pharmacodyn 2024; 51:581-604. [PMID: 38904912 PMCID: PMC11795844 DOI: 10.1007/s10928-024-09930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/07/2024] [Indexed: 06/22/2024]
Abstract
Quantitative predictive modeling of cancer growth, progression, and individual response to therapy is a rapidly growing field. Researchers from mathematical modeling, systems biology, pharmaceutical industry, and regulatory bodies, are collaboratively working on predictive models that could be applied for drug development and, ultimately, the clinical management of cancer patients. A plethora of modeling paradigms and approaches have emerged, making it challenging to compile a comprehensive review across all subdisciplines. It is therefore critical to gauge fundamental design aspects against requirements, and weigh opportunities and limitations of the different model types. In this review, we discuss three fundamental types of cancer models: space-structured models, ecological models, and immune system focused models. For each type, it is our goal to illustrate which mechanisms contribute to variability and heterogeneity in cancer growth and response, so that the appropriate architecture and complexity of a new model becomes clearer. We present the main features addressed by each of the three exemplary modeling types through a subjective collection of literature and illustrative exercises to facilitate inspiration and exchange, with a focus on providing a didactic rather than exhaustive overview. We close by imagining a future multi-scale model design to impact critical decisions in oncology drug development.
Collapse
Affiliation(s)
| | - Claire Couty
- Novadiscovery, 1 Place Giovanni Verrazzano, 69009, Lyon, France
| | - Paul Lemarre
- Novadiscovery, 1 Place Giovanni Verrazzano, 69009, Lyon, France
| | - Craig J Thalhauser
- Genmab US, Inc., 777 Scudders Mill Rd Bldg 2 4th Floor, Plainsboro, NJ, 08536, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
9
|
Cohen G, Rapoport B, Chan SW, Ruff P, Arance A, Mujika Eizmendi K, Houghton B, Brown MP, Zielinski RM, Muñoz Couselo E, Lyle M, Anderson JR, Jain L, de Alwis D, Lala M, Akala O, Chartash E, Jacobs C. Pembrolizumab 400 mg every 6 weeks as first-line therapy for advanced melanoma (KEYNOTE-555): Results from cohort B of an open-label, phase 1 study. PLoS One 2024; 19:e0309778. [PMID: 39531423 PMCID: PMC11556718 DOI: 10.1371/journal.pone.0309778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/02/2024] [Indexed: 11/16/2024] Open
Abstract
Intravenous pembrolizumab 400 mg every 6 weeks was approved across tumor types based on pharmacokinetic modeling, which showed exposures consistent with previous standard dosing of 200 mg or 2 mg/kg every 3 weeks, and early results of cohort B of the phase 1 KEYNOTE-555 study. Results after ≥1 year of potential follow-up for all patients in cohort B of KEYNOTE-555 are presented. Patients aged ≥18 years with previously untreated stage III/IV melanoma received pembrolizumab 400 mg every 6 weeks for ≤18 cycles. The primary endpoint was objective response rate per RECIST v1.1 by blinded independent central review. Secondary endpoints included duration of response, progression-free survival, pharmacokinetics, and safety. Overall, 101 patients received pembrolizumab. Median projected follow-up was 21.9 months (range, 17.0-25.7). The objective response rate was 50.5% (95% CI: 40.4-60.6; 19 complete responses, 32 partial responses). Median duration of response was not reached (NR; range, 2.4+ to 21.0+ months). Median progression-free survival was 13.8 months (95% CI: 4.1-NR). Observed pharmacokinetic exposures were consistent with model predictions for pembrolizumab 400 mg every 6 weeks and other approved and tested schedules (2 mg/kg or 200 mg every 3 weeks). Grade 3-4 treatment-related adverse events occurred in 13 patients (12.9%). No deaths were considered treatment related. These results support the pharmacokinetic modeling and demonstrate that the benefit-risk profile of pembrolizumab 400 mg Q6W is consistent with that of 200 mg or 2 mg/kg every 3 weeks. Clinically meaningful objective response rate and durable progression-free survival within the expected range for first-line pembrolizumab were observed. Clinical trial registry: ClinicalTrials.gov, NCT03665597.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Melanoma/drug therapy
- Melanoma/pathology
- Melanoma/mortality
- Middle Aged
- Female
- Male
- Aged
- Adult
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Aged, 80 and over
- Progression-Free Survival
- Cohort Studies
- Drug Administration Schedule
- Treatment Outcome
Collapse
Affiliation(s)
- Graham Cohen
- Mary Potter Oncology Centre, Pretoria, South Africa
| | - Bernardo Rapoport
- The Medical Oncology Centre of Rosebank, Johannesburg, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Sze W. Chan
- Sandton Oncology, Johannesburg, South Africa
| | - Paul Ruff
- University of Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Ana Arance
- Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Baerin Houghton
- Port Macquarie Base Hospital, Port Macquarie, New South Wales, Australia
| | - Michael P. Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, and School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert M. Zielinski
- Central West Cancer Care Centre, Orange Hospital, Orange, New South Wales, Australia
- Western Sydney University, Sydney, New South Wales, Australia
| | - Eva Muñoz Couselo
- Department of Medical Oncology, Melanoma and Other Skin Cancers Unit, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Megan Lyle
- Liz Plummer Cancer Care Centre, Cairns, Queensland, Australia
| | | | - Lokesh Jain
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Dinesh de Alwis
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Mallika Lala
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Omobolaji Akala
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Elliot Chartash
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Conrad Jacobs
- Cancer Care: Clinical & Radiation Oncology, Cape Town, South Africa
| |
Collapse
|
10
|
Nick C. Streamlining biosimilar development based on 20 years' experience. Expert Opin Biol Ther 2024; 24:571-581. [PMID: 38315062 DOI: 10.1080/14712598.2024.2314612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/01/2024] [Indexed: 02/07/2024]
Abstract
INTRODUCTION Biosimilar clinical programs could be streamlined by prudent application of improved methodologies and knowledge accumulated over the past 20 years. This review focuses on whether complex comparative efficacy trials are routinely needed and how to achieve a more tailored approach to biosimilar development. AREAS COVERED Key learnings over the past 20 years are summarized. It is noted that a one size fits all approach to biosimilar development is not appropriate: biological medicines fall within a wide spectrum of complexity, with blurring at the interface between biological products and small molecules. The interrelationship between quality, potency, pharmacokinetics, pharmacology, immunogenicity, efficacy, and safety are reviewed. Current regulatory thinking is reviewed with a look into what future challenges lie ahead. EXPERT OPINION To tailor regulatory requirements for marketing approval of biosimilars, it is proposed that a biosimilarity report be introduced. This report would integrate quality, pharmacology, immunogenicity, efficacy and safety findings and address how the clinical program could be tailored based on the totality of evidence.
Collapse
Affiliation(s)
- Cecil Nick
- Parexel International, Uxbridge, Middlesex, England
| |
Collapse
|
11
|
Li T, Ma W, Al-Obeidi E. Evolving Precision First-Line Systemic Treatment for Patients with Unresectable Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:2350. [PMID: 39001412 PMCID: PMC11240640 DOI: 10.3390/cancers16132350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
First-line systemic therapy for patients with advanced or metastatic non-small cell lung cancer (NSCLC) has rapidly evolved over the past two decades. First, molecularly targeted therapy for a growing number of gain-of-function molecular targets has been shown to improve progression-free survival (PFS) and overall survival (OS) with favorable toxicity profiles compared to platinum-containing chemotherapy and can be given as first-line systemic therapy in ~25% of patients with NSCLC. Actionable genetic alterations include EGFR, BRAF V600E, and MET exon 14 splicing site-sensitizing mutations, as well as ALK-, ROS1-, RET-, and NTRK-gene fusions. Secondly, inhibitors of programmed cell death protein 1 or its ligand 1 (PD-1/L1) such as pembrolizumab, atezolizumab, or cemiplimab monotherapy have become a standard of care for ~25% of patients with NSCLC whose tumors have high PD-L1 expression (total proportion score (TPS) ≥50%) and no sensitizing EGFR/ALK alterations. Lastly, for the remaining ~50% of patients who are fit and whose tumors have no or low PD-L1 expression (TPS of 0-49%) and no sensitizing EGFR/ALK aberrations, platinum-containing chemotherapy with the addition of a PD-1/L1 inhibitor alone or in combination of a cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitor improves PFS and OS compared to chemotherapy alone. The objectives of this review are to summarize the current data and perspectives on first-line systemic treatment in patients with unresectable NSCLC and propose a practical algorithm for implementing precision biomarker testing at diagnosis.
Collapse
Affiliation(s)
- Tianhong Li
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (W.M.)
- Medical Service, Hematology/Oncology, Veterans Affairs Northern California Health Care System, 10535 Hospital Way, Mather, CA 95655, USA
| | - Weijie Ma
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (W.M.)
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Ebaa Al-Obeidi
- Division of Hematology/Oncology, Department of Internal Medicine, University of California Davis School of Medicine, University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA; (W.M.)
| |
Collapse
|
12
|
Chen X, Ou S, Luo J, He Z, Jiang Q. Advancing perspectives on the off-label use of anticancer drugs: an updated classification and exploration of categories. Front Pharmacol 2024; 15:1374549. [PMID: 38898925 PMCID: PMC11186405 DOI: 10.3389/fphar.2024.1374549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
To date, the definition that the off-label usage of drugs refers to the unapproved use of approved drugs, which covers unapproved indications, patient populations, doses, and/or routes of administration, has been in existence for many years. Currently, there is a limited frequency and prevalence of research on the off-label use of antineoplastic drugs, mainly due to incomplete definition and classification issues. It is time to embrace new categories for the off-label usage of anticancer drugs. This review provided an insight into an updated overview of the concept and categories of the off-label use of anticancer drugs, along with illustrating specific examples to establish the next studies about the extent of the off-label usage of anticancer drugs in the oncology setting. The scope of the off-label use of current anticancer drugs beyond the previous definitions not only includes off-label uses in terms of indications, patient populations, doses, and/or routes of administration but also off-label use in terms of medication course, combination, sequence of medication, clinical purpose, contraindications scenarios, etc. In addition, the definition of the off-label usage of anticancer drugs should be added to the condition at a given time, and it varies from approval authorities. We presented a new and relatively comprehensive classification, providing extensive analysis and illustrative examples of the off-label usage of antineoplastic drugs for the first time. Such a classification has the potential to promote practical adoption and enhance management strategies for the off-label use of antitumor drugs.
Collapse
Affiliation(s)
- Xiaoyi Chen
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shunlong Ou
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jing Luo
- Department of Pharmacy, The Second People’s Hospital of Yibin, Yibin, Sichuan, China
| | - Zhisan He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Qian Jiang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Janssen JC, van Dijk B, de Joode K, Aarts MJB, van den Berkmortel FWPJ, Blank CU, Boers-Sonderen MJ, van den Eertwegh AJM, de Groot JWB, Jalving M, de Jonge MJA, Joosse A, Kapiteijn E, Kamphuis-Huismans AM, Naipal KAT, Piersma D, Rikhof B, Westgeest HM, Vreugdenhil G, Oomen-de Hoop E, Mulder EEAP, van der Veldt AAM. Safe Stop IPI-NIVO trial: early discontinuation of nivolumab upon achieving a complete or partial response in patients with irresectable stage III or metastatic melanoma treated with first-line ipilimumab-nivolumab - study protocol. BMC Cancer 2024; 24:632. [PMID: 38783238 PMCID: PMC11112744 DOI: 10.1186/s12885-024-12336-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Patients with irresectable stage III or metastatic melanoma presenting with poor prognostic factors are usually treated with a combination of immune checkpoint inhibitors (ICIs), consisting of ipilimumab and nivolumab. This combination therapy is associated with severe immune related adverse events (irAEs) in about 60% of patients. In current clinical practice, patients are usually treated with ICIs for up to two years or until disease progression or the occurrence of unacceptable AEs. The incidence of irAEs gradually increases with duration of treatment. While durable tumour responses have been observed after early discontinuation of treatment, no consensus has been reached on optimal treatment duration. The objective of the Safe Stop IPI-NIVO trial is to evaluate whether early discontinuation of ICIs is safe in patients with irresectable stage III or metastatic melanoma who are treated with combination therapy. METHODS The Safe Stop IPI-NIVO trial is a nationwide, multicentre, prospective, single-arm, interventional study in the Netherlands. A total of 80 patients with irresectable stage III or metastatic melanoma who are treated with combination therapy of ipilimumab-nivolumab and have a complete or partial response (CR/PR) according to RECIST v1.1 will be included to early discontinue maintenance therapy with anti-PD-1. The primary endpoint is the rate of ongoing response at 12 months after start of ICI. Secondary endpoints include ongoing response at 24 months, disease control at different time points, melanoma specific and overall survival, the incidence of irAEs and health-related quality of life. DISCUSSION From a medical, healthcare and economic perspective, overtreatment should be prevented and shorter treatment duration of ICIs is preferred. If early discontinuation of ICIs is safe for patients who are treated with the combination of ipilimumab-nivolumab, the treatment duration of nivolumab could be shortened in patients with a favourable tumour response. TRIAL REGISTRATION ClinicalTrials.gov ID NCT05652673, registration date: 08-12-2022.
Collapse
Affiliation(s)
- J C Janssen
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - B van Dijk
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - K de Joode
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - M J B Aarts
- Department of Medical Oncology, Maastricht UMC+, Maastricht, The Netherlands
| | | | - C U Blank
- Department of Medical Oncology, NKI-AvL, Amsterdam, The Netherlands
- Department of Medical Oncology, Leiden UMC, Leiden, The Netherlands
| | | | - A J M van den Eertwegh
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - J W B de Groot
- Department of Medical Oncology, Isala Zwolle, Zwolle, The Netherlands
| | - M Jalving
- Department of Medical Oncology, UMC Groningen, Groningen, The Netherlands
| | - M J A de Jonge
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - A Joosse
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - E Kapiteijn
- Department of Medical Oncology, Leiden UMC, Leiden, The Netherlands
| | | | - K A T Naipal
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - D Piersma
- Department of Medical Oncology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - B Rikhof
- Department of Medical Oncology, Medisch Centrum Leeuwarden, Leeuwarden, The Netherlands
| | - H M Westgeest
- Department of Medical Oncology, Amphia Ziekenhuis, Breda, The Netherlands
| | - G Vreugdenhil
- Department of Medical Oncology, Maxima Medisch Centrum Veldhoven, Veldhoven, The Netherlands
| | - E Oomen-de Hoop
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
| | - E E A P Mulder
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology and Radiology and Nuclear Medicine, Erasmus Medical Centre Cancer Institute, Dr. Molewaterplein 40, Rotterdam, 3015GD, The Netherlands.
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
14
|
Ji Y, Sy SKB. Utility and impact of quantitative pharmacology on dose selection and clinical development of immuno-oncology therapy. Cancer Chemother Pharmacol 2024; 93:273-293. [PMID: 38430307 DOI: 10.1007/s00280-024-04643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
Immuno-oncology (IO) therapies have changed the cancer treatment landscape. Immune checkpoint inhibitors (ICIs) have improved overall survival in 20-40% of patients with malignancies that were previously refractory. Due to the uniqueness in biology, modalities and patient responses, drug development strategies for IO differed from that traditionally used for cytotoxic and target therapies in oncology, and quantitative pharmacology utilizing modeling approach can be applied in all phases of the development process. In this review, we used case studies to showcase how various modeling methodologies were applied from translational science and dose selection through to label change, using examples that included anti-programmed-death-1 (anti-PD-1), anti-programmed-death ligand-1 (anti-PD-L1), anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), and anti-glucocorticoid-induced tumor necrosis factor receptor-related protein (anti-GITR) antibodies. How these approaches were utilized to support phase I-III dose selection, the design of phase III trials, and regulatory decisions on label change are discussed to illustrate development strategies. Model-based quantitative approaches have positively impacted IO drug development, and a better understanding of the biology and exposure-response relationship may benefit the development and optimization of new IO therapies.
Collapse
Affiliation(s)
- Yan Ji
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, 07936, USA.
| | - Sherwin K B Sy
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, 07936, USA.
| |
Collapse
|
15
|
Kao CH, Lin H, Liu CT, Ou YC, Fu HC, Wu CC, Wu CH. Real-world efficacy and safety of low-dose pembrolizumab in patients with advanced and refractory gynecologic cancers. J Formos Med Assoc 2024; 123:487-495. [PMID: 37852875 DOI: 10.1016/j.jfma.2023.09.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/31/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
OBJECTIVE The approved standard dose of pembrolizumab (200 mg administrated every 3 weeks) for cancer treatment imposes a significant financial burden on patients. However, no study has analyzed the clinical outcomes of low-dose pembrolizumab among individuals diagnosed with gynecologic cancer. The primary objective of this study was to assess the effectiveness and safety of a low-dose pembrolizumab regimen in real-world clinical practice. METHODS We retrospectively assessed the efficacy and safety data of patients with gynecologic malignancies who received pembrolizumab between 2017 and 2022 at Kaohsiung Chang Gung Memorial Hospital. Furthermore, we conducted a comparative analysis of the objective response rate (ORR) and progression-free survival (PFS) between patients with deficient mismatch repair (dMMR) and proficient MMR (pMMR). RESULTS A total of thirty-nine patients were included and received pembrolizumab at fixed dosages of 50 mg (5.1%), 100 mg (84.6%) and 200 mg (10.3%) per cycle. Compared to the pMMR group, the dMMR group exhibited a tendency toward improved ORR (45.5% vs. 13.0%, p = 0.074), and notably, the median duration of response remained unreached. There was no significant difference in PFS between the dMMR and pMMR groups; however, the patients with dMMR in tumor tissue had a trend of better survival (p = 0.079). Incidence of immune-related adverse events (irAEs) of any grade was observed in 13 patients (33.3%), with 3 individuals (7.7%) experiencing grade 3 or 4 events. CONCLUSION Low-dose pembrolizumab may be a cost-effective and safe treatment option without compromising clinical outcomes in patients with refractory gynecologic cancers.
Collapse
Affiliation(s)
- Chien-Hsiang Kao
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Ting Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Che Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Kaohsiung, Taiwan
| | - Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
16
|
Yu T, Liu X, Wu C, Tang Z, Wang H, Schnell P, Wan Y, Wang K, Liu L, Gao Y, Sahasranaman S, Budha N. Clinical dose rationale of tislelizumab in patients with solid or hematological advanced tumors. Clin Transl Sci 2024; 17:e13769. [PMID: 38515348 PMCID: PMC10958174 DOI: 10.1111/cts.13769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Tislelizumab, an anti-programmed cell death protein 1 monoclonal antibody, has demonstrated improved survival benefits over standard of care for multiple cancer indications. We present the clinical rationale and data supporting tislelizumab dose recommendation in patients with advanced tumors. The phase I, first-in-human, dose-finding BGB-A317-001 study (data cutoff [DCO]: August 2017) examined the following tislelizumab dosing regimens: 0.5-10 mg/kg every 2 weeks (q2w), 2-5 mg/kg q2w or q3w, and 200 mg q3w. Similar objective response rates (ORRs) were reported in the 2 and 5 mg/kg q2w or q3w cohorts. Safety outcomes (grade ≥3 adverse events [AEs], AEs leading to dose modification/discontinuation, immune-mediated AEs, and infusion-related reactions) were generally comparable across the dosing range examined. These results, alongside the convenience of a fixed q3w dose, formed the basis of choosing 200 mg q3w as the recommended dosing regimen for further clinical use. Pooled exposure-response (E-R) analyses by logistic regression using data from study BGB-A317-001 (DCO: August 2020) and three additional phase I/II studies (DCOs: 2018-2020) showed no statistically significant correlation between tislelizumab pharmacokinetic exposure and ORR across multiple solid tumor types or classical Hodgkin's lymphoma, nor was exposure associated with any of the safety end points evaluated over the dose range tested. Hence, tislelizumab showed a relatively flat E-R relationship. Overall, the totality of data, including efficacy, safety, and E-R analyses, together with the relative convenience of a fixed q3w dose, provided clinical rationale for the recommended dosing regimen of tislelizumab 200 mg q3w for multiple cancer indications.
Collapse
Affiliation(s)
- Tian Yu
- BeiGene USA, Inc.San MateoCaliforniaUSA
| | | | - Chi‐Yuan Wu
- BeiGene USA, Inc.San MateoCaliforniaUSA
- Present address:
Top Alliance Biosciences Inc.Menlo ParkCaliforniaUSA
| | | | | | | | - Ya Wan
- BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Kun Wang
- Shanghai Qiangshi Information Technology Co., Ltd.ShanghaiChina
| | - Lucy Liu
- Shanghai Qiangshi Information Technology Co., Ltd.ShanghaiChina
| | - Yuying Gao
- Shanghai Qiangshi Information Technology Co., Ltd.ShanghaiChina
| | - Srikumar Sahasranaman
- BeiGene USA, Inc.San MateoCaliforniaUSA
- Present address:
Prelude Therapeutics Inc.WilmingtonDelawareUSA
| | | |
Collapse
|
17
|
Guo Y, Remaily BC, Thomas J, Kim K, Kulp SK, Mace TA, Ganesan LP, Owen DH, Coss CC, Phelps MA. Antibody Drug Clearance: An Underexplored Marker of Outcomes with Checkpoint Inhibitors. Clin Cancer Res 2024; 30:942-958. [PMID: 37921739 PMCID: PMC10922515 DOI: 10.1158/1078-0432.ccr-23-1683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/23/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Immune-checkpoint inhibitor (ICI) therapy has dramatically changed the clinical landscape for several cancers, and ICI use continues to expand across many cancer types. Low baseline clearance (CL) and/or a large reduction of CL during treatment correlates with better clinical response and longer survival. Similar phenomena have also been reported with other monoclonal antibodies (mAb) in cancer and other diseases, highlighting a characteristic of mAb clinical pharmacology that is potentially shared among various mAbs and diseases. Though tempting to attribute poor outcomes to low drug exposure and arguably low target engagement due to high CL, such speculation is not supported by the relatively flat exposure-response relationship of most ICIs, where a higher dose or exposure is not likely to provide additional benefit. Instead, an elevated and/or increasing CL could be a surrogate marker of the inherent resistant phenotype that cannot be reversed by maximizing drug exposure. The mechanisms connecting ICI clearance, therapeutic efficacy, and resistance are unclear and likely to be multifactorial. Therefore, to explore the potential of ICI CL as an early marker for efficacy, this review highlights the similarities and differences of CL characteristics and CL-response relationships for all FDA-approved ICIs, and we compare and contrast these to selected non-ICI mAbs. We also discuss underlying mechanisms that potentially link mAb CL with efficacy and highlight existing knowledge gaps and future directions where more clinical and preclinical investigations are warranted to clearly understand the value of baseline and/or time-varying CL in predicting response to ICI-based therapeutics.
Collapse
Affiliation(s)
- Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Bryan C. Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Latha P. Ganesan
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Dwight H. Owen
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Mitch A. Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| |
Collapse
|
18
|
Xu S, Zhang N, Rinne ML, Sun H, Stein AM. Sabatolimab (MBG453) model-informed drug development for dose selection in patients with myelodysplastic syndrome/acute myeloid leukemia and solid tumors. CPT Pharmacometrics Syst Pharmacol 2023; 12:1653-1665. [PMID: 37186155 PMCID: PMC10681456 DOI: 10.1002/psp4.12962] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/16/2023] [Accepted: 03/20/2023] [Indexed: 05/17/2023] Open
Abstract
Sabatolimab is a novel immunotherapy with immuno-myeloid activity that targets T-cell immunoglobulin domain and mucin domain-3 (TIM-3) on immune cells and leukemic blasts. It is being evaluated for the treatment of myeloid malignancies in the STIMULUS clinical trial program. The objective of this analysis was to support the sabatolimab dose-regimen selection in hematologic malignancies. A population pharmacokinetic (PopPK) model was fit to patients with solid tumors and hematologic malignancies, which included acute myeloid leukemia, myelodysplastic syndrome (including intermediate-, high-, and very high-risk per Revised International Prognostic Scoring System), and chronic myelomonocytic leukemia. The PopPK model, together with a predictive model of sabatolimab distribution to the bone marrow and binding to TIM-3 was used to predict membrane-bound TIM-3 bone marrow occupancy. In addition, the total soluble TIM-3 (sTIM-3) kinetics and the pharmacokinetic (PK) exposure-response relationship in patients with hematologic malignancies were examined. At intravenous doses above 240 mg Q2w and 800 mg Q4w, we observed linear PK, a plateau in the accumulation of total sTIM-3, and a flat exposure-response relationship for both safety and efficacy. In addition, the model predicted membrane-bound TIM-3 occupancy in the bone marrow was above 95% in over 95% of patients. Therefore, these results support the selection of the 400 mg Q2w and 800 mg Q4w dosing regimens for the STIMULUS clinical trial program.
Collapse
Affiliation(s)
- Siyan Xu
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | - Na Zhang
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | | | - Haiying Sun
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | - Andrew M. Stein
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| |
Collapse
|
19
|
Lim K, Abegesah A, Fan C, He JZ, Song X, Chen C, Negro A, Makowsky M, Gupta C, Ren S, Phipps A, Gibbs M, Zhou D. Population Pharmacokinetics and Exposure-Response Analysis of Tremelimumab 300 mg Single Dose Combined with Durvalumab 1500 mg Q4W (STRIDE) in Patients with Unresectable Hepatocellular Carcinoma. J Clin Pharmacol 2023; 63:1221-1231. [PMID: 37300457 DOI: 10.1002/jcph.2288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023]
Abstract
A novel single-dose regimen of 300 mg tremelimumab in combination with durvalumab (STRIDE) has demonstrated a favorable benefit-risk profile in the phase 1/2 Study 22 trial (in patients with unresectable hepatocellular carcinoma, uHCC) and in the phase 3 HIMALAYA study. The current analysis evaluated the population pharmacokinetics (PopPK) of tremelimumab and durvalumab, and the exposure-response (ER) relationship for efficacy and safety of STRIDE in patients with uHCC. Previous PopPK models for tremelimumab and durvalumab were updated using data from previous studies in various cancers combined with data from Study 22 and HIMALAYA. Typical population mean parameters and associated inter- and intra-individual variability were assessed, as was the influence of covariates. Individual exposure metrics were derived from the individual empirical Bayes estimates as drivers for ER analysis related to efficacy and safety from HIMALAYA. The observed pharmacokinetics of tremelimumab in uHCC were well described by a 2-compartment model with both linear and time-dependent clearance. All identified covariates changed tremelimumab PK parameters by <25%, and thus had minimal clinical relevance; similar results were obtained from durvalumab PopPK analysis. None of tremelimumab or durvalumab exposure metrics were significantly associated with overall survival (OS), progression-free survival (PFS), or adverse events. Baseline aspartate aminotransferase and neutrophil-to-lymphocyte ratio (NLR) were associated with OS (P < .001) by the Cox proportional hazards model. No covariate was identified as a significant factor for PFS. No dose adjustment for tremelimumab or durvalumab is needed based on PopPK covariate analyses or ER analyses. Our findings support the novel STRIDE dosing regimen in patients with uHCC.
Collapse
Affiliation(s)
- KyoungSoo Lim
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Aburough Abegesah
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Chunling Fan
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Jimmy Zhijian He
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Xuyang Song
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Cecil Chen
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, South San Francisco, CA, USA
| | - Alejandra Negro
- Clinical Development, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Mallory Makowsky
- Clinical Development, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Charu Gupta
- Oncology Biometrics, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Song Ren
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Alex Phipps
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Megan Gibbs
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - Diansong Zhou
- Clinical Pharmacology & Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| |
Collapse
|
20
|
Turner DC, Wada R, Zhou H, Wang X, de Greef R, Valiathan C, Zhang L, Zhang N, Kuchimanchi M, Chen T, Ballas M, Visser SAG. Model-based meta-analysis of non-small cell lung cancer with standard of care PD-1 inhibitors and chemotherapy for early development decision making. CPT Pharmacometrics Syst Pharmacol 2023; 12:1751-1763. [PMID: 36642813 PMCID: PMC10681483 DOI: 10.1002/psp4.12917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/17/2023] Open
Abstract
Single-arm cohorts/trials are often used in early phase oncology programs to support preliminary clinical activity assessments for investigational products, administered alone or in combination with standard of care (SOC) agents. Benchmarking clinical activity of those combinations against other treatments, including SOC, requires indirect comparisons against external trials, which presents challenges including cross-study differences in trial populations/other factors. To facilitate such nonrandomized comparisons, we developed a comprehensive model-based meta-analysis (MBMA) framework to quantitatively adjust for factors related to efficacy in metastatic non-small cell lung cancer (mNSCLC). Data were derived from 15 published studies assessing key programmed cell death protein-1 (PD-1) inhibitors pembrolizumab (n = 8) and nivolumab (n = 7), representing current SOC in mNSCLC. In the first stage, a mixed-effects logistic regression model for overall response rate (ORR) was developed accounting for effects of various population covariates on ORR. The ORR model results indicated an odds ratio (OR) of 1.02 for squamous versus non-squamous histology and OR of 1.20 for PD-ligand 1 tumor proportion score (TPS) per every 10% increase of TPS level. Next, a nonparametric mixed-effects model for overall survival (OS) was developed with ORR/other clinical covariates as input. Subsequently, MBMA simulations of relevant hypothetical scenarios involving single-arm trial design predicted OS hazard ratios as a function of ORR with matched patient characteristics. Findings from this MBMA and derived parameter estimates can be generally applied by the reader as a framework for interpreting efficacy data from early phase trials to support ORR-based go/no-go decisions and futility rules, illustrated through examples in this report.
Collapse
Affiliation(s)
- David C. Turner
- GSKCollegevillePennsylvaniaUSA
- Present address:
GenentechSan FranciscoCaliforniaUSA
| | - Russ Wada
- CertaraMenlo ParkCaliforniaUSA
- Present address:
QuanTx ConsultingMountain ViewCaliforniaUSA
| | | | - Xiaowei Wang
- GSKCollegevillePennsylvaniaUSA
- Present address:
ModernaCambridgeMassachusettsUSA
| | | | - Chandni Valiathan
- GSKCollegevillePennsylvaniaUSA
- Present address:
J&JNew BrunswickNew JerseyUSA
| | | | | | | | | | - Marc Ballas
- GSKCollegevillePennsylvaniaUSA
- Present address:
NovocurePotomacMarylandUSA
| | | |
Collapse
|
21
|
Kondrashov A, Sapkota S, Sharma A, Riano I, Kurzrock R, Adashek JJ. Antibody-Drug Conjugates in Solid Tumor Oncology: An Effectiveness Payday with a Targeted Payload. Pharmaceutics 2023; 15:2160. [PMID: 37631374 PMCID: PMC10459723 DOI: 10.3390/pharmaceutics15082160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are at the forefront of the drug development revolution occurring in oncology. Formed from three main components-an antibody, a linker molecule, and a cytotoxic agent ("payload"), ADCs have the unique ability to deliver cytotoxic agents to cells expressing a specific antigen, a great leap forward from traditional chemotherapeutic approaches that cause widespread effects without specificity. A variety of payloads can be used, including most frequently microtubular inhibitors (auristatins and maytansinoids), as well as topoisomerase inhibitors and alkylating agents. Finally, linkers play a critical role in the ADCs' effect, as cleavable moieties that serve as linkers impact site-specific activation as well as bystander killing effects, an upshot that is especially important in solid tumors that often express a variety of antigens. While ADCs were initially used in hematologic malignancies, their utility has been demonstrated in multiple solid tumor malignancies, including breast, gastrointestinal, lung, cervical, ovarian, and urothelial cancers. Currently, six ADCs are FDA-approved for the treatment of solid tumors: ado-trastuzumab emtansine and trastuzumab deruxtecan, both anti-HER2; enfortumab-vedotin, targeting nectin-4; sacituzuzmab govitecan, targeting Trop2; tisotumab vedotin, targeting tissue factor; and mirvetuximab soravtansine, targeting folate receptor-alpha. Although they demonstrate utility and tolerable safety profiles, ADCs may become ineffective as tumor cells undergo evolution to avoid expressing the specific antigen being targeted. Furthermore, the current cost of ADCs can be limiting their reach. Here, we review the structure and functions of ADCs, as well as ongoing clinical investigations into novel ADCs and their potential as treatments of solid malignancies.
Collapse
Affiliation(s)
- Aleksei Kondrashov
- Department of Internal Medicine, Saint Agnes Hospital, Baltimore, MD 21229, USA; (A.K.); (S.S.)
| | - Surendra Sapkota
- Department of Internal Medicine, Saint Agnes Hospital, Baltimore, MD 21229, USA; (A.K.); (S.S.)
| | - Aditya Sharma
- Department of Internal Medicine, Dartmouth Health, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; (A.S.); (I.R.)
| | - Ivy Riano
- Department of Internal Medicine, Dartmouth Health, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA; (A.S.); (I.R.)
- Division of Hematology and Oncology, Dartmouth Cancer Center, Lebanon, NH 03755, USA
| | - Razelle Kurzrock
- WIN Consortium, 94550 Paris, France;
- MCW Cancer Center, Milwaukee, WI 53226, USA
- Division of Oncology and Hematology, University of Nebraska, Omaha, NE 68198, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Jacob J. Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
22
|
Haas AR, Golden RJ, Litzky LA, Engels B, Zhao L, Xu F, Taraszka JA, Ramones M, Granda B, Chang WJ, Jadlowsky J, Shea KM, Runkle A, Chew A, Dowd E, Gonzalez V, Chen F, Liu X, Fang C, Jiang S, Davis MM, Sheppard NC, Zhao Y, Fraietta JA, Lacey SF, Plesa G, Melenhorst JJ, Mansfield K, Brogdon JL, Young RM, Albelda SM, June CH, Tanyi JL. Two cases of severe pulmonary toxicity from highly active mesothelin-directed CAR T cells. Mol Ther 2023; 31:2309-2325. [PMID: 37312454 PMCID: PMC10422001 DOI: 10.1016/j.ymthe.2023.06.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/13/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023] Open
Abstract
Multiple clinical studies have treated mesothelin (MSLN)-positive solid tumors by administering MSLN-directed chimeric antigen receptor (CAR) T cells. Although these products are generally safe, efficacy is limited. Therefore, we generated and characterized a potent, fully human anti-MSLN CAR. In a phase 1 dose-escalation study of patients with solid tumors, we observed two cases of severe pulmonary toxicity following intravenous infusion of this product in the high-dose cohort (1-3 × 108 T cells per m2). Both patients demonstrated progressive hypoxemia within 48 h of infusion with clinical and laboratory findings consistent with cytokine release syndrome. One patient ultimately progressed to grade 5 respiratory failure. An autopsy revealed acute lung injury, extensive T cell infiltration, and accumulation of CAR T cells in the lungs. RNA and protein detection techniques confirmed low levels of MSLN expression by benign pulmonary epithelial cells in affected lung and lung samples obtained from other inflammatory or fibrotic conditions, indicating that pulmonary pneumocyte and not pleural expression of mesothelin may lead to dose-limiting toxicity. We suggest patient enrollment criteria and dosing regimens of MSLN-directed therapies consider the possibility of dynamic expression of mesothelin in benign lung with a special concern for patients with underlying inflammatory or fibrotic conditions.
Collapse
Affiliation(s)
- Andrew R Haas
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Ryan J Golden
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Leslie A Litzky
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Boris Engels
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Linlin Zhao
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Fangmin Xu
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - John A Taraszka
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Melissa Ramones
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Brian Granda
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Wan-Jung Chang
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Julie Jadlowsky
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kim-Marie Shea
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Adam Runkle
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Anne Chew
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Emily Dowd
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vanessa Gonzalez
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Fang Chen
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Xiaojun Liu
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chongyun Fang
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Shuguang Jiang
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Megan M Davis
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Neil C Sheppard
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Yangbing Zhao
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Simon F Lacey
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Gabriela Plesa
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - J Joseph Melenhorst
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Keith Mansfield
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | | | - Regina M Young
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven M Albelda
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Pulmonary, Allergy, and Critical Care, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Janos L Tanyi
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Liu D, Hu L, Shao H. Therapeutic drug monitoring of immune checkpoint inhibitors: based on their pharmacokinetic properties and biomarkers. Cancer Chemother Pharmacol 2023:10.1007/s00280-023-04541-8. [PMID: 37410155 DOI: 10.1007/s00280-023-04541-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/03/2023] [Indexed: 07/07/2023]
Abstract
As a new means of oncology treatment, immune checkpoint inhibitors (ICIs) can improve survival rates in patients with resistant or refractory tumors. However, there are obvious inter-individual differences in the unsatisfactory response rate, drug resistance rate and the occurrence of immune-related adverse events (irAE). These questions have sparked interest in researchers looking for a way to screen sensitive populations and predict efficacy and safety. Therapeutic drug monitoring (TDM) is a way to ensure the safety and effectiveness of medication by measuring the concentration of drugs in body fluids and adjusting the medication regimen. It has the potential to be an adjunctive means of predicting the safety and efficacy of ICIs treatment. In this review, the author outlined the pharmacokinetic (PK) characteristics of ICIs in patients. The feasibility and limitations of TDM of ICIs were discussed by summarizing the relationships between the pharmacokinetic parameters and the efficacy, toxicity and biomarkers.
Collapse
Affiliation(s)
- Dongxue Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Linlin Hu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
- Office of Medication Clinical Institution, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Hua Shao
- Office of Medication Clinical Institution, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
24
|
Faoro L, Brusegan A, Russi A, Calderone V, Martelli A, Marranconi E, Carpanese D, Berti E, Coppola M. Analysis of the relation between adverse events and overall survival in patients treated with pembrolizumab as a first-line treatment for metastatic NSCLC. BMC Pharmacol Toxicol 2023; 24:32. [PMID: 37189193 PMCID: PMC10184366 DOI: 10.1186/s40360-023-00663-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 03/14/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Many trials supported pembrolizumab as a first-line monotherapy to significantly improve overall survival (OS) in selected patients with previously untreated metastatic Non-Small Cell Lung Cancer (mNSCLC) and a PD-L1 TPS of ≥50% without EGFR/ALK mutations. The aim of this study was to reveal the correlation between OS and adverse events in real-world settings after 42 months. METHODS This retrospective observational study involved 98 patients with mNSCLC, TPS ≥ 50%, and no EGFR/ALK aberrations. Patients were treated with pembrolizumab (200 mg q3w) as a first-line treatment. Clinical data, including PD-L1 expression, Performance Status (ECOG-PS), treatment duration, toxicity, and outcomes were retrieved from local electronic medical records and from the Italian Regulatory Agency Registry. RESULTS The cohort's main characteristics were as follows: median age 73 [44-89] years, 64.3% were male and 35.7% were female, an ECOG-PS score of 0 (n = 73) and 1 or 2 (n = 25), and a PD-L1 > 90% in 29.6% of patients. The entire cohort had stage IV NSCLC at diagnosis. The median number of cycles was 8.5 at a median follow-up of 13 months. The median OS of 13.6 months (95% CI: 11.7-NA) was not influenced by sex and PD-L1, but was significantly associated with ECOG-PS (p = 0.02). Immune-Related Adverse Events (irAEs) occurred in 77.5% of patients (30.1% cutaneous, 27.5% gastrointestinal, and 20.4% endocrinological), but no grade 4 or 5 irAEs were identified. Patients experiencing any type of toxicity had a significantly longer median OS (20.39 months, 95% CI: 13.08-NA) than those with no toxicities (6.46 months, 95% CI: 1.41-NA, p = 0.006). CONCLUSION The percentage of irAEs detected was comparable to that reported in KEYNOTE-024 and KEYNOTE-042. These real-world findings demonstrated the significant correlation between OS and cutaneous toxicities.
Collapse
Affiliation(s)
- Lisa Faoro
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy.
| | - Adriana Brusegan
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| | - Alberto Russi
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Ettore Marranconi
- Department of Pharmacy, Specialization School in Hospital Pharmacy, University of Pisa, Pisa, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Elena Berti
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Marina Coppola
- Pharmacy Unit, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
25
|
Ren S, Feng J, Ma S, Chen H, Ma Z, Huang C, Zhang L, He J, Wang C, Zhou J, Danchaivijtr P, Wang CC, Vynnychenko I, Wang K, Orlandi F, Sriuranpong V, Li B, Ge J, Dang T, Zhou C. KEYNOTE-033: Randomized phase 3 study of pembrolizumab vs docetaxel in previously treated, PD-L1-positive, advanced NSCLC. Int J Cancer 2023. [PMID: 37141294 DOI: 10.1002/ijc.34532] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 05/05/2023]
Abstract
KEYNOTE-033 (NCT02864394) was a multicountry, open-label, phase 3 study that compared pembrolizumab vs docetaxel in previously treated, programmed death-ligand 1 (PD-L1)-positive, advanced non-small cell lung cancer (NSCLC), with most patients enrolled in mainland China. Eligible patients were randomized (1:1) to pembrolizumab 2 mg/kg or docetaxel 75 mg/m2 every 3 weeks. Primary endpoints were overall survival (OS) and progression-free survival and were evaluated sequentially using stratified log-rank tests, first in patients with PD-L1 tumor proportion score (TPS) ≥50% and then in patients with PD-L1 TPS ≥1% (significance threshold: P < .025, one-sided). A total of 425 patients were randomized to pembrolizumab (N = 213) or docetaxel (N = 212) between 8 September 2016 and 17 October 2018. In patients with a PD-L1 TPS ≥50% (n = 227), median OS was 12.3 months with pembrolizumab and 10.9 months with docetaxel; the hazard ratio (HR) was 0.83 (95% confidence interval [CI]: 0.61-1.14; P = .1276). Because the significance threshold was not met, sequential testing of OS and PFS was ceased. In patients with a PD-L1 TPS ≥1%, the HR for OS for pembrolizumab vs docetaxel was 0.75 (95% CI: 0.60-0.95). In patients from mainland China (n = 311) with a PD-L1 TPS ≥1%, HR for OS was 0.68 (95% CI: 0.51-0.89). Incidence of grade 3 to 5 treatment-related AEs was 11.3% with pembrolizumab vs 47.5% with docetaxel. In summary, pembrolizumab improved OS vs docetaxel in previously treated, PD-L1-positive NSCLC without unexpected safety signals; although the statistical significance threshold was not reached, the numeral improvement is consistent with that previously observed for pembrolizumab in previously treated, advanced NSCLC.
Collapse
Affiliation(s)
- Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jifeng Feng
- Oncology Department, Jiangsu Cancer Hospital, Nanjing, China
| | - Shenglin Ma
- Department of Thoracic Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Zhejiang University Cancer Center, Zhejiang, People's Republic of China
| | - HuaJun Chen
- Guangdong Provincial People's Hospital, Guangdong Lung Cancer Institute, Guangzhou, China
| | - Zhiyong Ma
- Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Zhengzhou, China
| | - Cheng Huang
- Department of Pneumology, Fujian Provincial Cancer Hospital, Fuzhou, China
| | - Li Zhang
- Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changli Wang
- Department of Biotherapy 17F, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Jianying Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, China
| | | | | | - Ihor Vynnychenko
- RMI Sumy Regional Clinical Oncology Dispensary, Sumy State University, Sumy, Ukraine
| | - Kai Wang
- Department of Respiration Medicine, The Second Affiliated Hospital of Zhejiang University College of Medicine, Hangzhou, China
| | - Francisco Orlandi
- Region Metropolitana de Santiago, Orlandi Oncologia, Providencia, Chile
| | - Virote Sriuranpong
- Medical Oncology Unit, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Ben Li
- MSD China, Beijing, China
| | - Jun Ge
- MSD China, Shanghai, China
| | | | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Dehghani T, Shahrjerdi A, Kahrizi MS, Soleimani E, Ravandeh S, Merza MS, Rahnama N, Ebrahimzadeh F, Bakhshesh M. Targeting programmed cell death protein 1 (PD-1) for treatment of non-small-cell lung carcinoma (NSCLC); the recent advances. Pathol Res Pract 2023; 246:154470. [PMID: 37150133 DOI: 10.1016/j.prp.2023.154470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/09/2023]
Abstract
The immune system uses various immune checkpoint axes to adjust responses, support homeostasis, and deter self-reactivity and autoimmunity. Nevertheless, non-small-cell lung carcinoma (NSCLC) can use protective mechanisms to facilitate immune evasion, which leads to potentiated cancer survival and proliferation. In this light, many blocking anti-bodies have been developed to negatively regulate checkpoint molecules, in particular, programmed cell death protein 1 (PD-1) / PD-ligand 1 (L1), and bypass these immune suppressive mechanisms. Meanwhile, anti-PD-1 anti-bodies such as nivolumab, pembrolizumab, cemiplimab, and sintilimab have shown excellent competence in successfully inspiring immune responses versus NSCLC. Accordingly, the United States Food and Drug Administration (FDA) has recently approved nivolumab (alone or in combination with ipilimumab) and pembrolizumab (alone or in combination with chemotherapy) as first-line treatment for advanced NSCLC patients. However, PD-1 blockade monotherapy remains inefficient in more than 60% of NSCLC patients, and many patients don't respond or acquire resistance to this modality. Also, toxicities related to anti-PD-1 anti-body have been progressively identified in clinical trials and oncology practice. Herein, we will outline the clinical benefits of PD-1 blockade therapy alone or in combination with other treatments (e.g., chemotherapy, radiotherapy, anti-angiogenic therapy) in NSCLC patients. Moreover, we will take a glimpse into the recently identified predictive biomarkers to determine patients most likely to suffer serious adverse events to decrease untoward toxicity risk and diminish treatment costs.
Collapse
Affiliation(s)
- Tannaz Dehghani
- Department of Internal Medicine, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Alireza Shahrjerdi
- National Institute for Genetic Engineering and Biotechnology (NIGEB), P.O. Box: 14965/161, Tehran, Iran
| | | | - Elnaz Soleimani
- Departmant of Genetic, Babol University of Medical Science, Babol, Iran
| | | | - Muna S Merza
- Prosthetic Dental Techniques Department, Al-Mustaqbal university College, Babylon 51001, Iraq
| | - Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Morteza Bakhshesh
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran.
| |
Collapse
|
27
|
Meriggi F, Zaniboni A, Zaltieri A. Low-Dose Immunotherapy: Is It Just an Illusion? Biomedicines 2023; 11:biomedicines11041032. [PMID: 37189650 DOI: 10.3390/biomedicines11041032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
The development and use of immunotherapy in the last decade have led to a drastic improvement in results in the onco-haematological field. This has implied, on the one hand, the need for clinicians to manage a new type of adverse event and, on the other hand, a significant increase in costs. However, emerging scientific evidence suggests that, as with other drugs in the recent past, the registry dosage can be drastically reduced for immunotherapies without penalizing their effectiveness. This would also lead to an important reduction in costs, expanding the audience of cancer patients who could access immunotherapy-based treatments. In this “Commentary”, we analyze the available evidence of pharmacokinetics and pharmacodynamics and the most recent literature in favor of low-dose immunotherapy.
Collapse
|
28
|
Lahiri A, Maji A, Potdar PD, Singh N, Parikh P, Bisht B, Mukherjee A, Paul MK. Lung cancer immunotherapy: progress, pitfalls, and promises. Mol Cancer 2023; 22:40. [PMID: 36810079 PMCID: PMC9942077 DOI: 10.1186/s12943-023-01740-y] [Citation(s) in RCA: 472] [Impact Index Per Article: 236.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/22/2022] [Indexed: 02/23/2023] Open
Abstract
Lung cancer is the primary cause of mortality in the United States and around the globe. Therapeutic options for lung cancer treatment include surgery, radiation therapy, chemotherapy, and targeted drug therapy. Medical management is often associated with the development of treatment resistance leading to relapse. Immunotherapy is profoundly altering the approach to cancer treatment owing to its tolerable safety profile, sustained therapeutic response due to immunological memory generation, and effectiveness across a broad patient population. Different tumor-specific vaccination strategies are gaining ground in the treatment of lung cancer. Recent advances in adoptive cell therapy (CAR T, TCR, TIL), the associated clinical trials on lung cancer, and associated hurdles are discussed in this review. Recent trials on lung cancer patients (without a targetable oncogenic driver alteration) reveal significant and sustained responses when treated with programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) checkpoint blockade immunotherapies. Accumulating evidence indicates that a loss of effective anti-tumor immunity is associated with lung tumor evolution. Therapeutic cancer vaccines combined with immune checkpoint inhibitors (ICI) can achieve better therapeutic effects. To this end, the present article encompasses a detailed overview of the recent developments in the immunotherapeutic landscape in targeting small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). Additionally, the review also explores the implication of nanomedicine in lung cancer immunotherapy as well as the combinatorial application of traditional therapy along with immunotherapy regimens. Finally, ongoing clinical trials, significant obstacles, and the future outlook of this treatment strategy are also highlighted to boost further research in the field.
Collapse
Affiliation(s)
- Aritraa Lahiri
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Avik Maji
- Department of Radiation Oncology, N. R. S. Medical College & Hospital, 138 A.J.C. Bose Road, Kolkata, 700014, India
| | - Pravin D Potdar
- Department of Molecular Medicine and Stem Cell Biology, Jaslok Hospital and Research Centre, Mumbai, 400026, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Purvish Parikh
- Department of Clinical Hematology, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, 302022, India
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, 400012, India
| | - Bharti Bisht
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Anubhab Mukherjee
- Esperer Onco Nutrition Pvt Ltd, 4BA, 4Th Floor, B Wing, Gundecha Onclave, Khairani Road, Sakinaka, Andheri East, Mumbai, Maharashtra, 400072, India.
| | - Manash K Paul
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
29
|
Song X, Kelley RK, Khan AA, Standifer N, Zhou D, Lim K, Krishna R, Liu L, Wang K, McCoon P, Negro A, He P, Gibbs M, Kurland JF, Abou-Alfa GK. Exposure-Response Analyses of Tremelimumab Monotherapy or in Combination with Durvalumab in Patients with Unresectable Hepatocellular Carcinoma. Clin Cancer Res 2023; 29:754-763. [PMID: 36477555 PMCID: PMC9932581 DOI: 10.1158/1078-0432.ccr-22-1983] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE A novel single-dose regimen of 300 mg tremelimumab in combination with durvalumab [Single Tremelimumab Regular Interval Durvalumab (STRIDE)] has demonstrated a favorable benefit-risk profile in the phase I/II Study 22 (NCT02519348) and phase III HIMALAYA study (NCT03298451). This study evaluated the pharmacokinetics, exposure-response, and exposure-pharmacodynamics relationships of tremelimumab in patients with unresectable hepatocellular carcinoma (uHCC). PATIENTS AND METHODS A previous tremelimumab population pharmacokinetic model was validated using data from parts 2 and 3 of Study 22. Exposure-response analyses explored relationships of tremelimumab exposure with efficacy and safety. Pharmacokinetics and pharmacodynamics relationships were evaluated using linear and nonlinear regression models. RESULTS The observed pharmacokinetics of tremelimumab in uHCC were consistent with predictions; no significant covariates were identified. Tremelimumab exposure was not significantly associated with adverse events, objective response rate, or progression-free survival. Overall survival (OS) was longer for patients with tremelimumab exposure, minimum serum drug concentration (Cmin1) ≥ median versus Cmin1 < median (18.99 months vs. 10.97 months), but this exposure-survival analysis might be confounded with baseline characteristics of albumin level and neutrophil to lymphocyte ratio, which had a significant impact on OS (P = 0.0004 and 0.0001, respectively). The predicted Cmin1 of tremelimumab in STRIDE regimen (12.9 μg/mL) was greater than the estimated concentration of tremelimumab eliciting half-maximal increases (EC50 = 5.24 μg/mL) in CD8+Ki67+ T-cell counts. CONCLUSIONS Our findings support novel insights into tremelimumab pharmacokinetics and exposure-response relationships in HCC and support the clinical utility of the STRIDE regimen in patients with uHCC.
Collapse
Affiliation(s)
- Xuyang Song
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Robin Kate Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California
| | - Anis A. Khan
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | | | - Diansong Zhou
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, Massachusetts
| | - KyoungSoo Lim
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - Rajesh Krishna
- Department of Clinical Pharmacology, Certara USA, Princeton, New Jersey
| | - Lu Liu
- Pharmacometrics, Shanghai Qiangshi Information Technology, Shanghai, China
| | - Kun Wang
- Pharmacometrics, Shanghai Qiangshi Information Technology, Shanghai, China
| | - Patricia McCoon
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, Massachusetts
| | | | - Philip He
- Oncology Biometrics, AstraZeneca, Gaithersburg, Maryland
| | - Megan Gibbs
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland
| | - John F. Kurland
- Immuno-oncology Franchise, Gastrointestinal and Head & Neck Cancers, AstraZeneca, Gaithersburg, Maryland
| | - Ghassan K. Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Medical College at Cornell University, New York, New York
| |
Collapse
|
30
|
Wang N, Zheng L, Li M, Hou X, Zhang B, Chen J, Li S, Chen L. Clinical efficacy and safety of individualized pembrolizumab administration based on pharmacokinetic in advanced non-small cell lung cancer: A prospective exploratory clinical trial. Lung Cancer 2023; 178:183-190. [PMID: 36868179 DOI: 10.1016/j.lungcan.2023.02.009] [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: 08/22/2022] [Revised: 11/24/2022] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
INTRODUCTION Pembrolizumab is recommended with a fixed dose of 200 mg 3-weekly. We performed this study to explore the clinical efficacy and safety of pharmacokinetic (PK)-guided pembrolizumab administration in advanced non-small cell lung cancer (NSCLC). METHODS In this prospective exploratory study, we enrolled advanced NSCLC patients in Sun Yat-Sen University Cancer Center. Eligible patients received pembrolizumab 200 mg 3-weekly with or without chemotherapy for four cycles, then for patients without progressive disease (PD), pembrolizumab was administrated in new dose-intervals according to steady state plasma-concentration (Css) of pembrolizumab until PD. We set the effective concentration (Ce) at 15 μg/ml and new dose-intervals (T) was calculated according to Css of pembrolizumab using following equation: Css × 21D = Ce (15 μg/ml) × T. Primary endpoint was the progression-free survival (PFS), secondary endpoints were objective response rate (ORR) and safety. Besides, advanced NSCLC patients received pembrolizumab 200 mg 3-weekly and more than four cycles in our center were defined as the history-controlled cohort. Patients with Css of pembrolizumab underwent genetic polymorphism analysis of variable number of tandem repeats (VNTR) region in neonatal Fc receptor (FcRn). The study was registered at ClinicalTrials.gov, NCT05226728. RESULTS A total 33 patients received pembrolizumab in new adjusted dose-intervals. The Css of pembrolizumab ranged from 11.01 to 61.21 μg/ml, 30 patients need prolonged intervals (22-80d) and 3 shortened intervals (15-20 d). In PK-guided cohort, the median PFS was 15.1 months and ORR 57.6 %, whereas in history-controlled cohort was 7.7 months and ORR 48.2 %. The immune-related adverse events were 15.2 % and 17.9 % between two cohort. The VNTR3/VNTR3 genotype of FcRn had significantly higher Css of pembrolizumab than VNTR2/VNTR3 (p = 0.005). CONCLUSIONS PK-guided pembrolizumab administration showed promising clinical efficacy and manageable toxicity. Meanwhile less frequent dosing of pembrolizumab by PK-guided could reduce financial toxicity potentially. This provided an alternative rational therapeutic strategy of pembrolizumab in advanced NSCLC.
Collapse
Affiliation(s)
- Na Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Lie Zheng
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Meichen Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Xue Hou
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Baishen Zhang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Jing Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Su Li
- Clinical Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| | - Likun Chen
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China.
| |
Collapse
|
31
|
Descourt R, Greillier L, Perol M, Ricordel C, Auliac JB, Falchero L, Gervais R, Veillon R, Vieillot S, Guisier F, Marcq M, Justeau G, Bigay-Game L, Bernardi M, Fournel P, Doubre H, Pinsolle J, Amrane K, Chouaïd C, Decroisette C. First-line single-agent pembrolizumab for PD-L1-positive (tumor proportion score ≥ 50%) advanced non-small cell lung cancer in the real world: impact in brain metastasis: a national French multicentric cohort (ESCKEYP GFPC study). Cancer Immunol Immunother 2023; 72:91-99. [PMID: 35729418 DOI: 10.1007/s00262-022-03232-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 05/25/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Few real-world data are available in patients with advanced metastatic non-small cell lung cancer (NSCLC) treated with first-line immunotherapy, particularly in those with brain metastases at treatment initiation. METHODS This was a national, retrospective, multicenter study that consecutively included all patients with PD-L1-positive (tumor proportion score ≥ 50%) advanced NSCLC who initiated first-line treatment with pembrolizumab as a single agent between May 2017 (date of availability of pembrolizumab in this indication in France) to November 22, 2019 (approval of the pembrolizumab-chemotherapy combination). Data were collected from medical records with local response assessment. RESULTS The cohort included 845 patients and 176 (20.8%) had brain metastases at diagnosis. There were no significant differences in outcomes for patients with and without brain metastases: 9.2 (95% CI 5.6-15) and 8 (95% CI 6.7-9.2, p = 0.3) months for median progression-free survival (PFS) and, 29.5 (95% CI 17.2-NA) and 22 (95% CI 17.8-27.1, p = 0.3) months for median overall survival (OS), respectively. Overall response rates were 47% and 45% in patients with and without cerebral metastases. In multivariate analysis, performance status 2-4 vs. 0-1 and neutrophil-to-lymphocyte ratio ≥ 4 vs. < 4 were the main independent negative factors for OS; brain metastasis was not an independent factor for OS. CONCLUSION In this large multicenter cohort, nearly 20% of patients initiating pembrolizumab therapy for advanced NSCLC had cerebral metastases. There was no significant difference in response rates, PFS and OS between patients with and without brain metastases.
Collapse
Affiliation(s)
- Renaud Descourt
- Oncology Department, Brest University Hospital, Brest, France
| | - Laurent Greillier
- Multidisciplinary Oncology and Therapeutic Innovations Department, APHM, INSERM, CNRS, CRCM, Hôpital Nord, Aix Marseille University, Marseille, France
| | - Maurice Perol
- Department of Medical Oncology, Léon Bérard Cancer Center, Lyon, France
| | - Charles Ricordel
- Pneumology Department, Rennes University Hospital, Rennes, France
| | - Jean-Bernard Auliac
- Pneumology Department, Creteil University Hospital, CHI Créteil, 40 Avenue de Verdun, 94010, Creteil, France
| | - Lionel Falchero
- Pneumology Department, Hôpital Nord-Ouest, Villefranche-sur-Saône Hospital, Gleize, France
| | - Radj Gervais
- Oncology Department, Caen François-Baclesse Cancer Center, Caen, France
| | - Rémi Veillon
- Pneumology Department, Bordeaux University Hospital, Bordeaux, France
| | | | - Florian Guisier
- Pneumology Department, Rouen University Hospital, Rouen, France
| | - Marie Marcq
- Pneumology Department, La Roche-sur-Yon Hospital, La Roche-sur-Yon, France
| | | | | | - Marie Bernardi
- Pneumology Department, Aix-en-Provence Hospital, Aix-en-Provence, France
| | - Pierre Fournel
- Oncology Department, Institut de Cancérologie de La Loire Lucien Neuwirth, Saint-Priest-en-Jarez, France
| | - Hélène Doubre
- Pneumology Department, Hôpital Foch, Suresnes, France
| | - Julian Pinsolle
- Pneumology Department, Chambéry Métropole Savoie Hospital, Chambéry, France
| | - Karim Amrane
- Oncology Department, Morlaix Hospital, Morlaix, France
| | - Christos Chouaïd
- Pneumology Department, Creteil University Hospital, CHI Créteil, 40 Avenue de Verdun, 94010, Creteil, France.
| | | |
Collapse
|
32
|
Clubb JHA, Kudling TV, Girych M, Haybout L, Pakola S, Hamdan F, Cervera-Carrascon V, Hemmes A, Grönberg-Vähä-Koskela S, Santos JM, Quixabeira DCA, Basnet S, Heiniö C, Arias V, Jirovec E, Kaptan S, Havunen R, Sorsa S, Erikat A, Schwartz J, Anttila M, Aro K, Viitala T, Vattulainen I, Cerullo V, Kanerva A, Hemminki A. Development of a Syrian hamster anti-PD-L1 monoclonal antibody enables oncolytic adenoviral immunotherapy modelling in an immunocompetent virus replication permissive setting. Front Immunol 2023; 14:1060540. [PMID: 36817448 PMCID: PMC9936529 DOI: 10.3389/fimmu.2023.1060540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer, but preclinical testing of hypotheses such as combination therapies has been complicated, in part due to species incompatibility issues. For example, one of few known permissive animal models for oncolytic adenoviruses is the Syrian hamster, for which an ICI, mainly an anti-PD-L1 monoclonal antibody (mAb) was not previously available. In this study, we developed an anti-Syrian hamster PD-L1 mAb to enable the evaluation of safety and efficacy, when combining anti-PD-L1 with an oncolytic adenovirus encoding tumour necrosis factor alpha (TNFα) and interleukin-2 (IL-2) (Ad5/3-E2F-D24-hTNFα-IRES-hIL-2 or TILT-123). Methods Recombinant Syrian hamster PD-L1 was expressed and mice immunized for mAb formation using hybridoma technology. Clonal selection through binding and functional studies in vitro, in silico and in vivo identified anti-PD-L1 clone 11B12-1 as the primary mAb candidate for immunotherapy modelling. The oncolytic virus (OV) and ICI combination approach was then evaluated using 11B12-1 and TILT-123 in a Syrian hamster model of pancreatic ductal adenocarcinoma (PDAC). Results Supernatants from hybridoma parent subclone 11B12B4 provided the highest positive PD-L1 signal, on Syrian hamster PBMCs and three cancer cell lines (HT100, HapT1 and HCPC1). In vitro co-cultures revealed superior immune modulated profiles of cell line matched HT100 tumour infiltrating lymphocytes when using subclones of 7G2, 11B12 and 12F1. Epitope binning and epitope prediction using AlphaFold2 and ColabFold revealed two distinct functional epitopes for clone 11B12-1 and 12F1-1. Treatment of Syrian hamsters bearing HapT1 tumours, with 11B12-1 induced significantly better (p<0.05) tumour growth control than isotype control by day 12. 12F1-1 did not induce significant tumour growth control. The combination of 11B12-1 with oncolytic adenovirus TILT-123 improved tumour growth control further, when compared to monotherapy (p<0.05) by day 26. Conclusions Novel Syrian hamster anti-PD-L1 clone 11B12-1 induces tumour growth control in a hamster model of PDAC. Combining 11B12-1 with oncolytic adenovirus TILT-123 improves tumour growth control further and demonstrates good safety and toxicity profiles.
Collapse
Affiliation(s)
- James H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Tatiana V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Lyna Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Firas Hamdan
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Víctor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Annabrita Hemmes
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Centre, Helsinki University Hospital, Helsinki, Finland
| | - João Manuel Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Elise Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Shreyas Kaptan
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Abdullah Erikat
- Department of Chemistry and the Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Joel Schwartz
- Chicago Department of Oral Medicine and Diagnostic Science, University of Illinois, Chicago, IL, United States
| | | | - Katri Aro
- Comprehensive Cancer Centre, Helsinki University Hospital, Helsinki, Finland.,Department of Otorhinolaryngology - Head and Neck Surgery, Helsinki Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tapani Viitala
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anna Kanerva
- Department of Gynecology and Obstetrics, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Centre, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
33
|
Qiu D, Xi H, Wang M, Jing P, Ren Z, Chang N, Jiang W, Yang X, Zhang Y, Chen X, Zhang Y, Zhang J. The debatable role of immune checkpoint blockade therapy in lung adenocarcinoma-oriented liver metastatic malignant lesions. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04538-5. [PMID: 36583744 DOI: 10.1007/s00432-022-04538-5] [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: 08/31/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND The use of anti-PD-1 or PD-L1 inhibitors in combination with other anti-cancer agents was a priority for treating advanced non-small cell lung cancer (NSCLC) patients with considerable PD-L1 expression. However, studies seldom show the progression of liver metastases after using immune checkpoint inhibitors (ICIs). METHODS Data were obtained from the Department of Pulmonary and Critical Care of Medicine, the First Affiliated Hospital of the Air Force Military Medical University. In the present study, we analyzed five non-small cell lung cancer (NSCLC) patients who had liver metastases after they were treated with pembrolizumab between 2019 and 2021. All of them had both stable primary lesions and liver progression with pembrolizumab intervention. Blood laboratory tests and imaging examinations were performed regularly during the treatment to assess the tumor responses of patients. RESULTS All patients displayed reduction or stability in the initial lesions as a result, but they also experienced the emergence of metastatic liver locations, which were regularly detected throughout immunotherapy. Additionally, the appearance of liver metastasis weakened their liver function gradually with the escalation of carcinoembryonic antigen, regarded as a predictor for evaluating the progression of tumors. These individuals were highly distinctive with hyper-progressive diseases associated with immunotherapy. We drew individualized intervention schemes for metastatic lesions in each patient and found that their life expectancy shared no significance given the restricting subjected population. CONCLUSIONS Our study indicated a clinical phenomenon after using immune checkpoint inhibitors and presented a necessity for implementing large scales clinical studies to manage NSCLC-oriented liver metastasis.
Collapse
Affiliation(s)
- Dan Qiu
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Hangtian Xi
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Min Wang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Pengyu Jing
- Department of Thoracic Surgery, The Second Affiliated Hospital of the Air Force Military Medical University, ShaanXi, China
| | - Zesheng Ren
- School of Basic Medicine, The Air Force Military Medical University, ShaanXi, China
| | - Ning Chang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Wenrui Jiang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Xuemin Yang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Yan Zhang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Xiangxiang Chen
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China
| | - Yong Zhang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China.
| | - Jian Zhang
- Department of Pulmonary and Critical Care of Medicine, The First Affiliated Hospital of the Air Force Military Medical University, #169 West Changle Road, Xi'an, 710032, ShaanXi Province, China.
| |
Collapse
|
34
|
Hobbs BP, Pestana RC, Zabor EC, Kaizer AM, Hong DS. Basket Trials: Review of Current Practice and Innovations for Future Trials. J Clin Oncol 2022; 40:3520-3528. [PMID: 35537102 PMCID: PMC10476732 DOI: 10.1200/jco.21.02285] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/06/2021] [Accepted: 03/31/2022] [Indexed: 02/05/2023] Open
Abstract
Advances in biology and immunology have elucidated genetic and immunologic origins of cancer. Innovations in sequencing technologies revealed that distinct cancer histologies shared common genetic and immune phenotypic traits. Pharmacologic developments made it possible to target these alterations, yielding novel classes of targeted agents whose therapeutic potential span multiple tumor types. Basket trials, one type of master protocol, emerged as a tool for evaluating biomarker-targeted therapies among multiple tumor histologies. Conventionally conducted within the phase II setting and designed to estimate high and durable objective responses, basket trials pose challenges to statistical design and interpretation of results. This article reviews basket trials implemented in oncology studies and discusses issues related to their statistical design and analysis.
Collapse
Affiliation(s)
- Brian P. Hobbs
- Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Roberto Carmagnani Pestana
- Centro de Oncologia e Hematologia Einstein Familia Dayan-Daycoval, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Emily C. Zabor
- Quantitative Health Sciences & Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Alexander M. Kaizer
- Biostatistics and Informatics, University of Colorado-Anschutz Medical Campus, Aurora, CO
| | - David S. Hong
- Investigational Cancer Therapeutics, University of Texas M.D. Anderson Cancer Center, Houston, TX
| |
Collapse
|
35
|
Melhem M, Hanze E, Lu S, Alskär O, Visser S, Gandhi Y. Population pharmacokinetics and exposure-response of anti-programmed cell death protein-1 monoclonal antibody dostarlimab in advanced solid tumours. Br J Clin Pharmacol 2022; 88:4142-4154. [PMID: 35357027 PMCID: PMC9543385 DOI: 10.1111/bcp.15339] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/03/2022] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
AIM Develop a population pharmacokinetic (PopPK) model to characterise the pharmacokinetics (PK) of anti-programmed cell death protein-1 (PD-1) antibody dostarlimab, identify covariates of clinical relevance, and investigate efficacy/safety exposure-response (ER) relationships. METHODS A PopPK model was developed using Phase 1 GARNET (NCT02715284) trial data for dostarlimab (1, 3 or 10 mg kg-1 every 2 wk; 500 mg every 3 wk or 1000 mg every 6 wk; 500 mg every 3 wk × 4 then 1000 mg every 6 wk [recommended regimen]) serum concentrations over time. Concentration-time data were analysed using nonlinear mixed effects modelling with standard stepwise covariate modelling. ER was explored for treatment-related adverse events and overall response rate (ORR) using logistic regression. RESULTS PopPK model/adverse event ER analyses included 546 patients (ORR ER analysis n = 362). Dostarlimab PK was well described by a 2-compartment model with time-dependent linear elimination. Time-dependent clearance decreased over time to a maximum of 14.9%. At steady state, estimated dostarlimab geometric mean coefficient of variation % clearance was 0.179 (30.2%) L d-1 ; volume of distribution was 5.3 (14.2%) L; terminal elimination half-life was 23.5 (22.4%) days. Statistically significant covariates were age, body weight, sex, time-varying albumin and alanine aminotransferase for clearance; body weight, albumin and sex for volume of distribution of the central compartment. Hepatic or renal impairment did not affect PK. There were no clinically significant ER relationships. CONCLUSION Dostarlimab PK parameters are similar to other anti-programmed cell death protein-1 antibodies. The clinical impact of covariates on exposure was limited-to-moderate, supporting recommended dostarlimab monotherapy therapeutic dosing.
Collapse
Affiliation(s)
| | | | - Sharon Lu
- GlaxoSmithKlineWalthamMAUSA
- Scholar RockCambridgeMAUSA
| | | | | | | |
Collapse
|
36
|
Füchsl F, Krackhardt AM. Paving the Way to Solid Tumors: Challenges and Strategies for Adoptively Transferred Transgenic T Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:4192. [PMID: 36077730 PMCID: PMC9454442 DOI: 10.3390/cancers14174192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 01/10/2023] Open
Abstract
T cells are important players in the antitumor immune response. Over the past few years, the adoptive transfer of genetically modified, autologous T cells-specifically redirected toward the tumor by expressing either a T cell receptor (TCR) or a chimeric antigen receptor (CAR)-has been adopted for use in the clinic. At the moment, the therapeutic application of CD19- and, increasingly, BCMA-targeting-engineered CAR-T cells have been approved and have yielded partly impressive results in hematologic malignancies. However, employing transgenic T cells for the treatment of solid tumors remains more troublesome, and numerous hurdles within the highly immunosuppressive tumor microenvironment (TME) need to be overcome to achieve tumor control. In this review, we focused on the challenges that these therapies must face on three different levels: infiltrating the tumor, exerting efficient antitumor activity, and overcoming T cell exhaustion and dysfunction. We aimed to discuss different options to pave the way for potent transgenic T cell-mediated tumor rejection by engineering either the TME or the transgenic T cell itself, which responds to the environment.
Collapse
Affiliation(s)
- Franziska Füchsl
- Klinik und Poliklinik für Innere Medizin III, School of Medicine, Technische Universität München, Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
| | - Angela M. Krackhardt
- Klinik und Poliklinik für Innere Medizin III, School of Medicine, Technische Universität München, Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
37
|
PD-L1 Over-Expression Varies in Different Subtypes of Lung Cancer: Will This Affect Future Therapies? Clin Pract 2022; 12:653-671. [PMID: 36136862 PMCID: PMC9498561 DOI: 10.3390/clinpract12050068] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 12/04/2022] Open
Abstract
Programmed death-ligand (PD-L) 1 and 2 are ligands of programmed cell death 1 (PD-1) receptor. They are members of the B7/CD28 ligand-receptor family and the most investigated inhibitory immune checkpoints at present. PD-L1 is the main effector in PD-1-reliant immunosuppression, as the PD-1/PD-L pathway is a key regulator for T-cell activation. Activation of T-cells warrants the upregulation of PD-1 and production of cytokines which also upregulate PD-L1 expression, creating a positive feedback mechanism that has an important role in the prevention of tissue destruction and development of autoimmunity. In the context of inadequate immune response, the prolonged antigen stimulation leads to chronic PD-1 upregulation and T-cell exhaustion. In lung cancer patients, PD-L1 expression levels have been of special interest since patients with non-small cell lung cancer (NSCLC) demonstrate higher levels of expression and tend to respond more favorably to the evolving PD-1 and PD-L1 inhibitors. The Food and Drug Administration (FDA) has approved the PD-1 inhibitor, pembrolizumab, alone as front-line single-agent therapy instead of chemotherapy in patients with NSCLC and PD-L1 ≥1% expression and chemoimmunotherapy regimens are available for lower stage disease. The National Comprehensive Cancer Network (NCCN) guidelines also delineate treatment by low and high expression of PD-L1 in NSCLC. Thus, studying PD-L1 overexpression levels in the different histological subtypes of lung cancer can affect our approach to treating these patients. There is an evolving role of immunotherapy in the other sub-types of lung cancer, especially small cell lung cancer (SCLC). In addition, within the NSCLC category, squamous cell carcinomas and non-G12C KRAS mutant NSCLC have no specific targetable therapies to date. Therefore, assessment of the PD-L1 expression level among these subtypes of lung cancer is required, since lung cancer is one of the few malignances wherein PD-L1 expression levels is so crucial in determining the role of immunotherapy. In this study, we compared PD-L1 expression in lung cancer according to the histological subtype of the tumor.
Collapse
|
38
|
Mayawala K, de Alwis D. Dose Finding in Oncology: What is Impeding Coming of Age? Pharm Res 2022; 39:1817-1822. [PMID: 35474158 PMCID: PMC9314272 DOI: 10.1007/s11095-022-03263-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022]
Abstract
After a drug molecule enters clinical trials, there are primarily three levers to enhance probability of success: patient selection, dose selection and choice of combination agents. Of these, dose selection remains an under-appreciated aspect in oncology drug development despite numerous peer-reviewed publications. Here, we share practical challenges faced by the biopharmaceutical industry that reduce the willingness to invest in dose finding for oncology drugs. First, randomized dose finding admittedly slows down clinical development. To reduce the size of dose finding study, trend in exposure vs. tumor-size analysis can be assessed, instead of a statistical test for non-inferiority between multiple doses. Second, investment in testing a lower dose when benefit-risk at the higher dose is sufficient for regulatory approval (i.e., efficacy at the higher dose is better than standard of care and safety is acceptable) is perceived as low priority. Changing regulatory landscape must be considered to optimize dose in pre-marketing setting as post-marketing changes in dose can be commercially costly. Third, the risk of exposing patients to subtherapeutic exposures with a lower dose should be assessed scientifically instead of assuming a monotonic relationship between dose and efficacy. Only the doses which are expected to be at the plateau of dose/exposure-response curve should be investigated in Phase 1b/2. Overall, changing the perceptions that have been impeding investment in dose finding in oncology requires pragmatic discourse among biopharmaceutical industry, regulatory agencies and academia. These perceptions should also not deter dose finding for recently emerging modalities, including BITEs and CART cell therapies.
Collapse
Affiliation(s)
- Kapil Mayawala
- Oncology Early Development, Clinical Research, Merck and Co., Inc., NJ, Kenilworth, USA.
| | - Dinesh de Alwis
- Oncology Early Development, Clinical Research, Merck and Co., Inc., NJ, Kenilworth, USA
| |
Collapse
|
39
|
余 孟, 王 洪. [Application of MIDD in Clinical Research of Antitumor Drugs]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:487-492. [PMID: 35899446 PMCID: PMC9346158 DOI: 10.3779/j.issn.1009-3419.2022.101.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 11/05/2022]
Abstract
The antitumor drug has become one of the focused areas in new drug research and development. Their clinical research generally consumes a long period of time, with high cost and high risk. Model-informed drug development (MIDD) integrates and quantitatively analyzes physiological, pharmacological, and disease progression information through modeling and simulation, which can reduce the cost of drug development and improve the efficiency of clinical research. In this essay, Osimertinib and Pembrolizumab are given as examples to illustrate the specific application of MIDD in different phases of clinical research, aiming to provide references for the application of MIDD to guide the clinical research of antitumor drugs.
.
Collapse
Affiliation(s)
- 孟洋 余
- />100730 北京,中国医学科学院北京协和医学院,北京协和医院,临床药理研究中心Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - 洪允 王
- />100730 北京,中国医学科学院北京协和医学院,北京协和医院,临床药理研究中心Clinical Pharmacology Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
40
|
Guo B, Zang Y. A Bayesian phase I/II biomarker-based design for identifying subgroup-specific optimal dose for immunotherapy. Stat Methods Med Res 2022; 31:1104-1119. [PMID: 35191780 PMCID: PMC9305985 DOI: 10.1177/09622802221080753] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunotherapy is an innovative treatment that enlists the patient's immune system to battle tumors. The optimal dose for treating patients with an immunotherapeutic agent may differ according to their biomarker status. In this article, we propose a biomarker-based phase I/II dose-finding design for identifying subgroup-specific optimal dose for immunotherapy (BSOI) that jointly models the immune response, toxicity, and efficacy outcomes. We propose parsimonious yet flexible models to borrow information across different types of outcomes and subgroups. We quantify the desirability of the dose using a utility function and adopt a two-stage dose-finding algorithm to find the optimal dose for each subgroup. Simulation studies show that the BSOI design has desirable operating characteristics in selecting the subgroup-specific optimal doses and allocating patients to those optimal doses, and outperforms conventional designs.
Collapse
Affiliation(s)
- Beibei Guo
- Department of Experimental Statistics, 5779Louisiana State University, Baton Rouge, USA
| | - Yong Zang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, USA
- Center for Computational Biology and Bioinformatics, Indiana University, Indianapolis, USA
| |
Collapse
|
41
|
Pembrolizumab Weight-Based Dosing: Conviction and Lacunae in Adopting a Cost-Saving Approach—A Survey Report. Indian J Med Paediatr Oncol 2022. [DOI: 10.1055/s-0042-1745814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
Abstract
Introduction Use of immunotherapy drugs has increased leaps and bounds in the last decade with promising results in some of the cancers. The use is limited in low- and middle-income countries due to cost constraints. Weight-based dosing is one measure adopted by Canada and Israel to reduce cost burden and improve access to immunotherapeutic drugs.
Objective We conducted a survey among medical oncologists from India to understand challenges faced in accepting the weight-based dosing of pembrolizumab.
Materials and Methods Questionnaire covering various aspects related to use of immunotherapy drugs was made and it was circulated across various social media platforms. Medical oncologists practicing across India were invited to participate in this survey. The issues like access to drugs and awareness about weight-based dosing of pembrolizumab were covered in the survey. Also, the impact of international guidelines on accepting the weigh-based dosing was studied.
Results Ninety-nine medical oncologists across India participated in the survey. Only 60% medical oncologists are aware about weight-based dosing of pembrolizumab practiced in other countries. Further, 70% of medical oncologists could not prescribe immunotherapy due to cost factor in majority (90%) of their patients. More than 90% agreed that they will use weight-based dosing of pembrolizumab if the Drug Controller General of India, National Comprehensive Cancer Network, or European Society of Medical Oncologists guidelines endorses weight-based dosing.
Conclusion Weight-based dosing of pembrolizumab would be accepted if policy makers and Indian medical oncology societies come together and formulate guidelines. Such guidelines will improve accessibility to immunotherapy drugs and lead to huge cost savings.
Collapse
|
42
|
Modified-Dose Pembrolizumab and Prognostic Outcomes among Non-Small Cell Lung Cancer Patients: A Chart Review Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19105999. [PMID: 35627534 PMCID: PMC9141635 DOI: 10.3390/ijerph19105999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023]
Abstract
The modified dose (MD) regimen of pembrolizumab (2 mg/kg or 100 mg every 3 weeks) is an alternative option to reduce the financial burden resulting from the extremely high cost of the standard dose (SD) regimen (200 mg every 3 weeks). However, the clinical effectiveness and prognostic outcomes have not been fully elucidated in real-word clinical practice. Sixty-four consecutive patients in Taiwan receiving pembrolizumab for advanced NSCLC between 2018 and 2020 were recruited in this study. Comparisons of overall survival (OS) and progression-free survival (PFS) were performed using Kaplan−Meier survival curves. Additionally, 12 predictors, including pembrolizumab regimen, dose, neutrophil-to-lymphocyte ratio (NLR), age, sex, histopathology, smoking history, ECOG PS, EGFR mutation, PD-L1 expression, distant metastases and treatment line, were analyzed in multivariable Cox models for predicting OS and PFS. The results showed that the MD group and the SD group had similar OS and PFS, especially in patients beyond first-line treatment or with a pretreatment NLR < 5. The NLR was the only independent factor associated with both OS (adjusted HR = 0.052; p = 0.010) and PFS (adjusted HR = 0.259; p = 0.021). The results of this study assure the clinical effectiveness of MD pembrolizumab and suggest that the pretreatment NLR could highlight patients who may benefit from MD pembrolizumab.
Collapse
|
43
|
Felip E, Moreno V, Morgensztern D, Curigliano G, Rutkowski P, Trigo JM, Calvo A, Kowalski D, Cortinovis D, Plummer R, Maio M, Ascierto PA, Vladimirov VI, Cervantes A, Zudaire E, Hazra A, T'jollyn H, Bandyopadhyay N, Greger JG, Attiyeh E, Xie H, Calvo E. First-in-human, open-label, phase 1/2 study of the monoclonal antibody programmed cell death protein-1 (PD-1) inhibitor cetrelimab (JNJ-63723283) in patients with advanced cancers. Cancer Chemother Pharmacol 2022; 89:499-514. [PMID: 35298698 PMCID: PMC8956549 DOI: 10.1007/s00280-022-04414-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 02/16/2022] [Indexed: 12/19/2022]
Abstract
Purpose To assess the safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of cetrelimab (JNJ-63723283), a monoclonal antibody programmed cell death protein-1 (PD-1) inhibitor, in patients with advanced/refractory solid tumors in the phase 1/2 LUC1001 study. Methods In phase 1, patients with advanced solid tumors received intravenous cetrelimab 80, 240, 460, or 800 mg every 2 weeks (Q2W) or 480 mg Q4W. In phase 2, patients with melanoma, non-small-cell lung cancer (NSCLC), and microsatellite instability–high (MSI-H)/DNA mismatch repair-deficient colorectal cancer (CRC) received cetrelimab 240 mg Q2W. Response was assessed Q8W until Week 24 and Q12W thereafter. Results In phase 1, 58 patients received cetrelimab. Two dose-limiting toxicities were reported and two recommended phase 2 doses (RP2D) were defined (240 mg Q2W or 480 mg Q4W). After a first dose, mean maximum serum concentrations (Cmax) ranged from 24.7 to 227.0 µg/mL; median time to Cmax ranged from 2.0 to 3.2 h. Pharmacodynamic effect was maintained throughout the dosing period across doses. In phase 2, 146 patients received cetrelimab 240 mg Q2W. Grade ≥ 3 adverse events (AEs) occurred in 53.9% of patients. Immune-related AEs (any grade) occurred in 35.3% of patients (grade ≥ 3 in 6.9%). Overall response rate was 18.6% across tumor types, 34.3% in NSCLC, 52.6% in programmed death ligand 1–high (≥ 50% by immunohistochemistry) NSCLC, 28.0% in melanoma, and 23.8% in centrally confirmed MSI-H CRC. Conclusions The RP2D for cetrelimab was established. Pharmacokinetic/pharmacodynamic characteristics, safety profile, and clinical activity of cetrelimab in immune-sensitive advanced cancers were consistent with known PD-1 inhibitors. Trial registrations NCT02908906 at ClinicalTrials.gov, September 21, 2016; EudraCT 2016–002,017-22 at clinicaltrialsregister.eu, Jan 11, 2017. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04414-6.
Collapse
Affiliation(s)
- Enriqueta Felip
- Thoracic Cancer Unit, Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Victor Moreno
- Phase 1 Trials Unit, START MADRID-FJD, Hospital Fundación Jiménez Díaz Medical Oncology Division, Madrid, Spain
| | - Daniel Morgensztern
- Division of Oncology, Section of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Giuseppe Curigliano
- Division of Early Drug Development, European Institute of Oncology, IRCCS and University of Milano, Milan, Italy
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - José Manuel Trigo
- Department of Medical Oncology, Hospital Universitario Virgen de La Victoria y Regional, Malaga, Spain
| | - Aitana Calvo
- Oncology Service, Hospital General Universitario Gregorio Maranon, Madrid, Spain
| | - Dariusz Kowalski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Skłodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Ruth Plummer
- Sir Bobby Robson Unit, Northern Centre for Cancer Care, Newcastle Hospitals NHS Trust and Newcastle University, Newcastle, UK
| | - Michele Maio
- Center for Immuno-Oncology, Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy
| | - Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS-Fondazione Pascale, Napoli, Italy
| | | | - Andres Cervantes
- Medical Oncology Department, INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
| | | | - Anasuya Hazra
- Janssen Research & Development, Spring House, PA, USA
| | | | | | | | | | - Hong Xie
- Janssen Research & Development, Spring House, PA, USA
| | - Emiliano Calvo
- Centro Integral Oncológico Clara Campal Medical Oncology Division, START Madrid-CIOCC, Sanchinarro University Hospital, Madrid, Spain.
| |
Collapse
|
44
|
Patel A, Akhade A, Parikh P, Sharma A, Malhotra H, Prabhash K, Babu G, Noronha V, Batra U, Mehta P, Gupta VG, Radhakrishnan V, Boya RR, Biswas B. Pembrolizumab weight based dosing – A call for policy change. Indian J Med Paediatr Oncol 2022. [DOI: 10.1055/s-0042-1742651] [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] Open
Affiliation(s)
- Amol Patel
- Department of Medicine, Oncology Centre, INHS, ASVINI, Mumbai
| | - Amol Akhade
- Department of Medical Oncology, Topiwala National Medical College and Nair Hospital, Mumbai, Maharashtra, India
| | - Purvish Parikh
- Department of Medical Oncology, Mumbai Oncocare Centers, Mumbai, Maharashtra, India
| | - Atul Sharma
- Department of Medical Oncology, AIIMS, New Delhi, India
| | - Hemant Malhotra
- Department of Medical Oncology, Mahatmma Gandhi Medical College Hospital, Jaipur, Rajasthan, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Govind Babu
- Department of Medical Oncology, HCG and St John’s Medical College Hospital, Bengaluru, Karnataka, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Ullas Batra
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Delhi, India
| | - Prashant Mehta
- Department of Medical Oncology, Asian Institute of Medical Sciences, Faridabad, Haryana, India
| | | | | | - Rakesh Reddy Boya
- Department of Medical Oncology, Mahatma Gandhi Cancer Hospital, Vishakhapatnam, Andhra Pradesh, India
| | - Bivas Biswas
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| |
Collapse
|
45
|
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
|
46
|
Homšek A, Radosavljević D, Miletić N, Spasić J, Jovanović M, Miljković B, Stanojković T, Vučićević K. Review of the Clinical Pharmacokinetics, Efficacy and Safety of Pembrolizumab. Curr Drug Metab 2022; 23:460-472. [PMID: 35692130 DOI: 10.2174/1389200223666220609125013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/13/2022] [Accepted: 03/11/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Treatment of various types of cancer has been improved significantly with the discovery of biological drugs that act as immune checkpoint inhibitors (ICIs). Pembrolizumab is a humanized monoclonal anti- PD-1 antibody currently approved for the treatment of a wide range of tumors, with more indications still being investigated in ongoing clinical trials. OBJECTIVE The aim of this paper is to present all currently available data regarding pembrolizumab pharmacokinetic and pharmacodynamic characteristics. Also, the possibility of using predictive biomarkers to monitor patients during cancer treatment is discussed. METHODS Database research was carried out (PubMed, ScienceDirect). Information was gathered from original articles, the European Medicines Agency datasheets and results from clinical trials. RESULTS This review summarizes present-day knowledge about the pharmacokinetics, different modeling approaches and dosage regimens, efficacy and safety of pembrolizumab and therapeutic monitoring of disease progression. CONCLUSION This review points out consistent pharmacokinetic characteristics of pembrolizumab in various cancer patients, the lack of pharmacokinetic-pharmacodynamic/outcome relationships, and the need for adequate biomarkers to predict treatment success. Hence, there is a clear necessity for more data and experience in order to optimize pembrolizumab treatment for each individual patient.
Collapse
Affiliation(s)
- Ana Homšek
- Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Davorin Radosavljević
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Nebojša Miletić
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jelena Spasić
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Marija Jovanović
- Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Branislava Miljković
- Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Tatjana Stanojković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Katarina Vučićević
- Department of Pharmacokinetics and Clinical Pharmacy, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
47
|
Rubio-Viqueira B, Tarruella MM, Lázaro M, Estévez SV, Córdoba-Ortega JF, Maiques IM, González JG, Cordellat AB, Valdivia-Bautista J, Arenas CG, Sánchez Torres JM. PD-L1 testing and clinical management of newly diagnosed metastatic non-small cell lung cancer in Spain: MOREL study. Lung Cancer Manag 2021; 10:LMT53. [PMID: 34899993 PMCID: PMC8656292 DOI: 10.2217/lmt-2021-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/04/2021] [Indexed: 11/21/2022] Open
Abstract
Aim To describe the clinical management and PD-L1 testing of patients with newly diagnosed stage IV non-small cell lung cancer (NSCLC) without driver mutations in Spain. Methods Multicenter, retrospective study. Results Among 297 evaluated patients, 89.2% received systemic treatment for stage IV disease, of whom 53.6% received platinum doublet therapy, 26.8% immunotherapy as monotherapy and 14.7% immunotherapy + chemotherapy, with 9.4% receiving treatment as part of a clinical trial. Treatment was initiated 1 month after histological diagnosis, with PD-L1 test results available in most cases (92.6%). PD-L1 testing was performed in 287 patients, 95.1% by in-house tests, mostly with the 22C3 pharmDx assay. The factor most strongly associated with treatment selection was, as expected, the expression of PD-L1. Conclusion PD-L1 testing is implemented in clinical practice and seems to guide treatment decisions in patients with NSCLC in Spain.
Collapse
Affiliation(s)
- Belen Rubio-Viqueira
- Department of Medical Oncology, Quirónsalud Madrid Hospital, Pozuelo de Alarcón, Madrid, Spain
| | | | - Martín Lázaro
- Department of Medical Oncology, Álvaro Cunqueiro University Hospital, Vigo, Pontevedra, 36213, Spain
| | | | | | | | - Jorge García González
- Department of Medical Oncology, University Clinical Hospital of Santiago de Compostela, A Coruña, 15706, Spain
| | - Ana Blasco Cordellat
- Department of Medical Oncology, Hospital General Universitario de Valencia, CIBERONC, Valencia, 46014, Spain
| | | | | | | |
Collapse
|
48
|
Mayawala K, Nayak T, Jain L, de Alwis D. Mechanistic Basis for Maximally Efficacious Dose of Pembrolizumab. Clin Pharmacol Ther 2021; 111:994. [PMID: 34859427 DOI: 10.1002/cpt.2492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022]
Affiliation(s)
| | - Tapan Nayak
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Lokesh Jain
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | |
Collapse
|
49
|
Wong PY, How SH, Hassan R, Abdul Azih MN. Limited doses of immunotherapy use in advanced non-small cell lung cancer elderly patients with ECOG of 2 and high PDL-1 expression. Aging Med (Milton) 2021; 4:345-349. [PMID: 34964016 PMCID: PMC8711218 DOI: 10.1002/agm2.12179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/30/2021] [Accepted: 10/08/2021] [Indexed: 12/05/2022] Open
Abstract
Immunotherapy is an effective treatment in advanced non-small cell lung cancer (NSCLC) patients with high PDL-1 expression. Here, we report three such patients with durable response despite limited doses immunotherapy.
Collapse
Affiliation(s)
- Pui Yee Wong
- Department of Respiratory MedicineHospital Tengku Ampuan AfzanKuantanMalaysia
| | - Soon Hin How
- Department of MedicineKulliyyah of MedicineInternational Islamic University MalaysiaKuantanMalaysia
| | - Radhiana Hassan
- Department of RadiologyKulliyyah of MedicineInternational Islamic University MalaysiaKuantanMalaysia
| | | |
Collapse
|
50
|
Affiliation(s)
- Ian F. Tannock
- Division of Medical Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | - Amol Patel
- Department of Medicine, Oncology Centre, Indian Naval Hospital Ship, Asvini, Colaba, Mumbai, Maharashtra, India
| |
Collapse
|