1
|
Hughes DJ, Chand G, Johnson J, Tegala R, Bailey D, Adamson K, Edmonds S, Meszaros LK, Moore AEB, Manickavasagar T, Ndagire S, Gennatas S, Georgiou A, Ghosh S, Josephs D, Karapanagiotou E, McLean E, Ting HH, Spicer J, Goh V, Cook GJR. PD-L1 imaging with [ 99mTc]NM-01 SPECT/CT is associated with metabolic response to pembrolizumab with/without chemotherapy in advanced lung cancer. Br J Cancer 2025; 132:913-921. [PMID: 40188291 DOI: 10.1038/s41416-025-02991-w] [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: 08/08/2024] [Revised: 02/15/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) immunohistochemistry is a predictive biomarker for anti-PD-(L)1 therapy in non-small cell lung cancer (NSCLC). It is not a reliable predictor of clinical benefit with non-invasive imaging providing a potential solution. We present the PECan study, the aim of which to assess the relationship of [99mTc]-labeled anti-PD-L1 single-domain antibody (NM-01) single-photon emission computed tomography (SPECT)/CT with metabolic response to anti-PD-(L)1. METHODS PD-L1 tumour proportion score (TPS) measured using SP263 assay. [99mTc]NM-01 SPECT/CT and [18F]FDG PET/CT performed before and 9-weeks following pembrolizumab with/without chemotherapy in patients with advanced NSCLC. Tumor (T) to blood pool (BP) maximum region of interest (ROImax) measurements performed in primary and metastatic lesions using SPECT/CT images. RESULTS Fifteen patients were included (median age 63 years, 9 male). Intertumoural heterogeneity evident in 10(67%) patients. Mean [99mTc]NM-01 T:BP demonstrated moderate correlation with PD-L1 TPS (r = 0.45, p < 0.05). Depth of [18F]FDG PET/CT metabolic response at 9-weeks (n = 13), correlated strongly with baseline [99mTc]NM-01 T:BP (r = -0.73, p < 0.05), but only moderately with PD-L1 TPS (r = -0.46, p = 0.06). CONCLUSION [99mTc]NM-01 SPECT/CT allows non-invasive quantification of PD-L1 in primary tumour and metastases in NSCLC. [99mTc]NM-01 uptake moderately correlates with PD-L1 immunohistochemistry, determines heterogeneity, and is associated with early metabolic response to anti-PD-1 pembrolizumab. CLINICAL TRIALS REGISTRATION PD-L1 Expression in Cancer (PECan) study (NCT04436406), registered 18 June 2020 https://clinicaltrials.gov/ct2/show/NCT04436406.
Collapse
Affiliation(s)
- Daniel Johnathan Hughes
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gitasha Chand
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - Jessica Johnson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Ronan Tegala
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Damion Bailey
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Kathryn Adamson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | - Scott Edmonds
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, Guy's Hospital, London, UK
| | | | - Amelia Elizabeth Broomfield Moore
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
| | - Thubeena Manickavasagar
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Susan Ndagire
- King's Health Partners Cancer Biobank, Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Spyridon Gennatas
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Alexandros Georgiou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Sharmistha Ghosh
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Debra Josephs
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Eleni Karapanagiotou
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Emma McLean
- Department of Histopathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Hong Hoi Ting
- Nanomab Technology (UK) Limited, Borehamwood, Hertfordshire, UK
| | - James Spicer
- Cancer Centre at Guy's, Guy's and St Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, Great Maze Pond, London, UK
| | - Vicky Goh
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK
- Department of Radiology, Guy's and St. Thomas' NHS Foundation Trust, St Thomas' Hospital, London, UK
| | - Gary J R Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, 5th Floor Becket House, London, UK.
- King's College London & Guy's and St. Thomas' PET Centre, St Thomas' Hospital, London, UK.
| |
Collapse
|
2
|
Chen M, Zhou Y, Bao K, Chen S, Song G, Wang S. Multispecific Antibodies Targeting PD-1/PD-L1 in Cancer. BioDrugs 2025; 39:427-444. [PMID: 40106158 DOI: 10.1007/s40259-025-00712-6] [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: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The development of immune checkpoint inhibitors has revolutionized the treatment of patients with cancer. Targeting the programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1(PD-L1) interaction using monoclonal antibodies has emerged as a prominent focus in tumor therapy with rapid advancements. However, the efficacy of anti-PD-1/PD-L1 treatment is hindered by primary or acquired resistance, limiting the effectiveness of single-drug approaches. Moreover, combining PD-1/PD-L1 with other immune drugs, targeted therapies, or chemotherapy significantly enhances response rates while exacerbating adverse reactions. Multispecific antibodies, capable of binding to different epitopes, offer improved antitumor efficacy while reducing drug-related side effects, serving as a promising therapeutic approach in cancer treatment. Several bispecific antibodies (bsAbs) targeting PD-1/PD-L1 have received regulatory approval, and many more are currently in clinical development. Additionally, tri-specific antibodies (TsAbs) and tetra-specific antibodies (TetraMabs) are under development. This review comprehensively explores the fundamental structure, preclinical principles, clinical trial progress, and challenges associated with bsAbs targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Yuli Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kaicheng Bao
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Siyu Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Guoqing Song
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| |
Collapse
|
3
|
Jiang C, Ren F, Zhang M, Lu Q, Zeng S, Yang G, Zhu Y. Using Pharmacokinetic and Pharmacodynamic Analysis to Optimize the Dosing Regimens of Fanastomig (EMB-02) in Patients With Advanced Solid Tumors. CPT Pharmacometrics Syst Pharmacol 2025; 14:975-986. [PMID: 40067130 PMCID: PMC12072225 DOI: 10.1002/psp4.70011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 05/14/2025] Open
Abstract
Fanastomig (also known as EMB-02) is a bispecific antibody targeting programmed cell death protein-1(PD-1) and lymphocyte activation gene-3 (LAG-3), developed for the treatment of advanced solid tumors. A first-in-human (FIH) Phase I study (NCT04618393) evaluated safety, tolerability, pharmacokinetics (PK), pharmacodynamics (PD), immunogenicity, and clinical efficacy of Fanastomig in patients with advanced solid tumors. To determine the recommended Phase II dose (RP2D), population pharmacokinetics (PopPK), and exposure and response analysis (E-R) were conducted. The PopPK model, demonstrating good performance, showed no clinically meaningful relationship between areas under the concentration-time curve (AUC) or maximum concentration (Cmax) of Fanastomig and selected covariates of interest. A nonlinear Emax model was fitted to Fanastomig PD-1 receptor occupancy (RO) in the peripheral blood compartment. The estimated half-maximal effective concentration (EC50) was 0.084 μg/mL (95% confidence interval [CI]: 0.0369-0.131). Assuming a threefold lower exposure in tumor tissue compared to that in serum, a target trough concentration of Fanastomig at ~2.27 μg/mL would be needed for 90% PD-1 RO in the tumor. Modeling and simulation indicated that a weekly dosing (QW) of 360 mg would achieve full peripheral blood RO in approximately 90% of patients. The incidence of anti-drug antibodies (ADAs) for Fanastomig was high (95.7%, 44/46), with a negative correlation between the ADA titer and dose levels; meanwhile, ADA minimally impacted PK exposure and efficacy. An inverse trend was observed between anaphylaxis and PK exposure. Fanastomig was well tolerated and had acceptable safety profiles up to 900 mg QW. Based on these findings, two dosing regimens have been selected for further clinical development. Trial Registration: ClinicalTrials.gov identifier: NCT04618393.
Collapse
MESH Headings
- Humans
- Neoplasms/drug therapy
- Male
- Middle Aged
- Female
- Aged
- Adult
- Antibodies, Bispecific/administration & dosage
- Antibodies, Bispecific/pharmacokinetics
- Antibodies, Bispecific/pharmacology
- Antibodies, Bispecific/adverse effects
- Dose-Response Relationship, Drug
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Models, Biological
- Immune Checkpoint Inhibitors/administration & dosage
- Immune Checkpoint Inhibitors/pharmacokinetics
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/pharmacology
- Area Under Curve
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/pharmacology
Collapse
Affiliation(s)
- Chengjun Jiang
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| | - Fang Ren
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| | - Mingfei Zhang
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| | - Qiaoyang Lu
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| | - Shuqi Zeng
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| | - Guang Yang
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| | - Yonghong Zhu
- Hanghai EpimAb Biotherapeutics Co., LtdShanghai EpimAb BiotherapeuticsShanghaiChina
| |
Collapse
|
4
|
Yu T, Wu C, Sahasranaman S, Tian X, Fei Li Y, Tang Z, Yang Y, Wan Y, Zhang Q, Schnell P, Mendoza‐Naranjo A, Abdrashitov R, Hanley WD, Budha N. Clinical Pharmacology Overview of Tislelizumab in Patients With Advanced Tumors With a Focus on Racial Impact. Clin Transl Sci 2025; 18:e70221. [PMID: 40286322 PMCID: PMC12033005 DOI: 10.1111/cts.70221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/30/2025] [Accepted: 02/22/2025] [Indexed: 04/29/2025] Open
Abstract
Tislelizumab, an anti-programmed cell death protein-1 monoclonal antibody, has demonstrated improved survival over the standard of care for multiple cancers. However, tislelizumab's effectiveness across different racial/ethnicity groups warrants further evaluation. This clinical pharmacology overview includes tislelizumab's pharmacokinetic properties, correlations with efficacy and safety, and immunogenicity, with a focus on racial impact. Non-compartmental pharmacokinetic analysis was conducted using data from Asian and White patients enrolled in BGB-A317-001 and BGB-A317-102. Population pharmacokinetic analyses used pooled data from 12 clinical studies to evaluate the impact of intrinsic/extrinsic factors on tislelizumab's pharmacokinetic properties, including race effect. Exposure-efficacy/exposure-safety relationships and immunogenicity assessments were evaluated for the phase III BGB-A317-302/-303 studies. Tislelizumab exhibited dose-proportional pharmacokinetics, and there were no clinically meaningful differences in tislelizumab's pharmacokinetic parameters at 200 mg once every 3 weeks between BGB-A317-001 (n = 12, 83% White patients) and BGB-A317-102 (n = 20, 100% Chinese patients); race was not a significant covariate. No clinically relevant exposure-efficacy/-safety relationships were observed in BGB-A317-302/-303. Incidence of anti-drug antibodies (ADAs) was similar between Asian and White patients. The presence of ADAs was not clinically relevant for tislelizumab's pharmacokinetic properties, efficacy, or safety. There were no differences in tislelizumab's pharmacokinetic or ADA characteristics between Asian and White patients with advanced cancer and no clinically relevant exposure-efficacy/-safety dependency or impact of immunogenicity on efficacy and safety. Data from the extensive clinical program of tislelizumab support the use of tislelizumab across broad patient populations with relevant tumor types.
Collapse
MESH Headings
- Female
- Humans
- Male
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Asian People
- Clinical Trials, Phase III as Topic
- Dose-Response Relationship, Drug
- Neoplasms/drug therapy
- Neoplasms/ethnology
- Neoplasms/immunology
- Neoplasms/pathology
- Treatment Outcome
- White People
Collapse
Affiliation(s)
- Tian Yu
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Chi‐Yuan Wu
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | | | - Xianbin Tian
- PK SciencesNovartis Biomedical Research, Novartis Pharmaceutical, Inc.East HanoverNew JerseyUSA
| | - Ying Fei Li
- Pharmacometrics, Oncology, Global Drug DevelopmentNovartis Pharmaceutical, Inc.East HanoverNew JerseyUSA
| | - Zhiyu Tang
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Yanfei Yang
- Clinical BA BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Ya Wan
- Scientific Programming, BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Quting Zhang
- Scientific Programming, BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Patrick Schnell
- Global Product SafetyBeiGene USA, Inc.Ridgefield ParkNew JerseyUSA
| | | | | | - William D. Hanley
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Nageshwar Budha
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| |
Collapse
|
5
|
Shah M, Noronha V, Rajamanickam Kulandaivel S, Poladia B, Niyogi D, Menon N, Kaushal R, Shetty O, Pai T, Tibdewal A, Vora M, Shah D, Vora D, Shah S, Goud S, Shah A, Maske K, Shetake A, Prabhash K. Neoadjuvant Chemotherapy and Low Dose Immunotherapy in Resectable Non-small Cell Lung Cancer: A Multi-center Retrospective Cohort Analysis. Clin Oncol (R Coll Radiol) 2025; 41:103795. [PMID: 40073678 DOI: 10.1016/j.clon.2025.103795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Affiliation(s)
- M Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - V Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - S Rajamanickam Kulandaivel
- Department of Surgical Oncology, Thangam Cancer Center and Research Institute, Namakkal, Tamil Nadu, India
| | - B Poladia
- Department of Medical Oncology, Thangam Cancer Center and Research Institute, Namakkal, Tamil Nadu, India
| | - D Niyogi
- Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - N Menon
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - R Kaushal
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - O Shetty
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - T Pai
- Department of Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - A Tibdewal
- Department of Radiation Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - M Vora
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - D Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - D Vora
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - S Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - S Goud
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - A Shah
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - K Maske
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - A Shetake
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| | - K Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute, Dr E Borges Road, Parel, Mumbai 400012, Maharashtra, India
| |
Collapse
|
6
|
Marron TU, Luke JJ, Hoffner B, Perlmutter J, Szczepanek C, Anagnostou V, Silk AW, Romero PJ, Garrett-Mayer E, Emens LA. A SITC vision: adapting clinical trials to accelerate drug development in cancer immunotherapy. J Immunother Cancer 2025; 13:e010760. [PMID: 40121030 PMCID: PMC11931932 DOI: 10.1136/jitc-2024-010760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/03/2025] [Indexed: 03/25/2025] Open
Abstract
Clinical trials of cancer immunotherapy (IO) were historically based on a drug development paradigm built for chemotherapies. The remarkable clinical activity of programmed cell death protein 1/programmed death ligand 1 blockade, chimeric antigen receptor-T cells, and T cell engagers yielded new insights into how the mechanistic underpinnings of IO are reflected in the clinic. These insights and the sheer number of novel immunotherapies currently in the pipeline have made it clear that our strategies and tools for IO drug development must adapt. Recent innovations like engineered T cells and tumor-infiltrating lymphocytes demonstrate that immune-based treatments may rely on real-time manufacturing programs rather than off-the-shelf drugs. We now recognize adoptively transferred cells as living drugs. Progression criteria have been redefined due to the unique response patterns of IO. Harnessing the power of both biomarkers and the neoadjuvant setting earlier in drug development is of broad interest. The US Food and Drug Association is increasingly impacting the design of trials with respect to dose optimization and clinical endpoints. The use of novel endpoints such as pathologic complete/major response, treatment-free survival, and minimal residual disease is becoming more common. There is growing acceptance of using patient-reported outcomes as trial endpoints to better measure the true clinical benefit and impact of novel IO agents on quality of life. New opportunities created by modern data science and artificial intelligence to inform and accelerate drug development continue to emerge. The importance of streamlining the clinical research ecosystem and enhancing clinical trial access to facilitate the enrollment of diverse patient populations is broadly recognized. Patient advocacy is critical both to drive the science of IO, and to promote patient satisfaction. To capitalize on these opportunities, the Society for Immunotherapy of Cancer (SITC) has established a goal of at least 100 new, unique IO approvals over the next 10 years. Accordingly, SITC has developed initiatives designed to integrate the viewpoints of diverse stakeholders and galvanize the field in further adapting clinical trials to the unique features of IO, moving us closer to our ultimate goal of using IO to cure and prevent cancer.
Collapse
Affiliation(s)
- Thomas U Marron
- Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jason J Luke
- UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Brianna Hoffner
- Advanced Practitioner Society for Hematology and Oncology, Hightstown, New Jersey, USA
| | | | - Connie Szczepanek
- Cancer Research Consortium of West Michigan (CRCWM), Grand Rapids, Michigan, USA
| | - Valsamo Anagnostou
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ann W Silk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Elizabeth Garrett-Mayer
- Center for Research and Analytics, American Society of Clinical Oncology, Alexandria, Virginia, USA
| | | |
Collapse
|
7
|
Remon J, Bortolot M, Bironzo P, Cortiula F, Menis J, Brandao M, Naidoo J, van Geel R, Reguart N, Arrieta O, Mountzios G, Hendriks LEL, Besse B. De-Escalation Strategies With Immune Checkpoint Blockers in Non-Small Cell Lung Cancer: Do We Already Have Enough Evidence? J Clin Oncol 2025; 43:1148-1156. [PMID: 39836933 DOI: 10.1200/jco-24-02347] [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: 10/20/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 01/23/2025] Open
Abstract
Immune checkpoint blockers (ICBs) have revolutionized the treatment of non-small cell lung cancer (NSCLC). Currently, one-dose-fits-all maximalist regimens have been considered the standard of care, with ICBs administered at flat doses regardless of patients' weight. Treatment duration with ICBs is often arbitrary across stages, ranging from a fixed time point to until disease progression or unacceptable toxicity. However, the pharmacokinetic and pharmacodynamic properties of ICBs differ significantly from those of traditional cytotoxic drugs and the approved and selected doses on the basis of the maximum tolerated dose are often overestimated as there is limited evidence supporting a direct relationship between therapeutic intensity and outcomes. This can lead to overtreatment of patients, resulting in an increased risk of toxicity without enhanced efficacy. In addition, the use of these drugs is associated with significant costs that burden the global health care system and exacerbate disparities in access to care. De-escalating treatment by reducing the dose, duration, and frequency of administration of ICBs could optimize treatment efficacy, reduce toxicities, improve patients' quality of life, and even decrease costs. Ultimately, de-escalation strategies may help to reduce treatment inequalities and to improve drug access worldwide. The aim of this review is to summarize and discuss the main issues and challenges regarding the de-escalation of ICBs in patients with NSCLC, focusing on dose-intensity reduction and treatment duration selection. Moreover, we assess the economic impact of implementing de-escalation approaches.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Martina Bortolot
- Department of Pulmonary Diseases, Maastricht University Medical Centre+, GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Paolo Bironzo
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Oncology, University of Turin, Turin, Italy
| | - Francesco Cortiula
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre (+), GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
- University Hospital of Udine, Department of Oncology, Udine, Italy
| | - Jessica Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - Mariana Brandao
- Institute Jules Bordet-Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Jarushka Naidoo
- Beaumont Hospital and RCSI University of Health Sciences, Dublin, Ireland
| | - Robin van Geel
- Department of Clinical Pharmacy & Toxicology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Noemi Reguart
- Department of Oncology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, Maastricht University Medical Centre+, GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
8
|
Buehning F, Lerchner T, Vogel J, Hendgen-Cotta UB, Totzeck M, Rassaf T, Michel L. Preclinical models of cardiotoxicity from immune checkpoint inhibitor therapy. Basic Res Cardiol 2025; 120:171-185. [PMID: 39039301 PMCID: PMC11790694 DOI: 10.1007/s00395-024-01070-0] [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: 03/05/2024] [Revised: 06/30/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy represents a ground-breaking paradigm in cancer treatment, harnessing the immune system to combat malignancies by targeting checkpoints such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1). The use of ICI therapy generates distinctive immune-related adverse events (irAEs) including cardiovascular toxicity, necessitating targeted research efforts. This comprehensive review explores preclinical models dedicated to ICI-mediated cardiovascular complications including myocarditis. Tailored preclinical models of ICI-mediated myocardial toxicities highlight the key role of CD8+ T cells, emphasizing the profound impact of immune checkpoints on maintaining cardiac integrity. Cytokines and macrophages were identified as possible driving factors in disease progression, and at the same time, initial data on possible cardiac antigens responsible are emerging. The implications of contributing factors including thoracic radiation, autoimmune disorder, and the presence of cancer itself are increasingly understood. Besides myocarditis, mouse models unveiled an accelerated progression of atherosclerosis, adding another layer for a thorough understanding of the diverse processes involving cardiovascular immune checkpoint signalling. This review aims to discuss current preclinical models of ICI cardiotoxicity and their potential for improving enhanced risk assessment and diagnostics, offering potential targets for innovative cardioprotective strategies. Lessons from ICI therapy can drive novel approaches in cardiovascular research, extending insights to areas such as myocardial infarction and heart failure.
Collapse
Affiliation(s)
- Florian Buehning
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tobias Lerchner
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Julia Vogel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
9
|
Proctor JR, Wong H. Clinical study design strategies to mitigate confounding effects of time-dependent clearance on dose optimization of therapeutic antibodies. CPT Pharmacometrics Syst Pharmacol 2025; 14:365-375. [PMID: 39575837 PMCID: PMC11812930 DOI: 10.1002/psp4.13280] [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: 09/10/2024] [Accepted: 10/29/2024] [Indexed: 02/13/2025] Open
Abstract
Time-dependent pharmacokinetics (TDPK) is a frequent confounding factor that misleads exposure-response (ER) analysis of therapeutic antibodies, where a decline in clearance results in increased drug exposure over time in patients who respond to therapy, causing a false-positive ER finding. The object of our simulation study was to explore the influence of clinical trial designs on the frequency of false-positive ER findings. Two previously published population PK models representative of slow- (pembrolizumab) and fast-onset (rituximab) TDPK were used to simulate virtual patient cohorts with time-dependent clearance and the frequency of false-positive ER findings. The impact of varying the number of dose groups, dose range, and sample size was evaluated over time. Study designs with a single tested dose level showed a high probability of showing a false-positive ER finding. When TDPK has a slow onset, use of exposure measures from early timepoints in ER analysis significantly reduces the risk of a false-positive, while with fast onset it did not. Randomization of patients to two dose levels greatly reduced the risk, with a threefold or greater dose range offering the greatest benefit. The likelihood of false-positive increases with a larger sample size, where greater care should be taken to identify confounding factors. Clinical trial simulation supports that appropriate clinical study design and analysis with adequate dose exploration can reduce but cannot entirely eliminate the risk of misleading ER findings.
Collapse
Affiliation(s)
- Jeffrey R. Proctor
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Harvey Wong
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
10
|
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
|
11
|
Tamuli B, Ghagare R, Mandal G. Design, Production, and Optimization of Antigen-Specific Recombinant Antitumor Dimeric IgA Antibody. Methods Mol Biol 2025; 2909:119-129. [PMID: 40029519 DOI: 10.1007/978-1-0716-4442-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Available cancer immunotherapies are currently restricted to extracellular targets, while chemotherapy is the only option for intracellular targets, such as mutant KRAS. Antibodies are serum immunoglobulins, each having high binding specificity against particular antigens. Patients produce antibody responses against abnormally expressed self-proteins and neoantigens presented by the cancer cells. However, despite their infiltration into the tumor beds, many times the magnitude of the antitumor antibodies produced by spontaneously infiltrated B lymphocytes remains insufficient to control tumor growth. Recent work has established that dimeric IgA antibodies can target intracellular targets inside cancer cells expressing the polymeric immunoglobulin receptor (pIgR). Here, we thoroughly discuss the entire process of recombinant production of intracellular antigen-specific dimeric IgA antibodies that could be utilized for targeting oncodrivers inside tumor cells.
Collapse
Affiliation(s)
- Baishali Tamuli
- Cancer Immune Environment and Therapeutics Lab, Tumor Immunology and Immunotherapy, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Rutik Ghagare
- Division of Cancer Biology, BRIC-Institute of Life Sciences, Bhubaneswar, India
| | - Gunjan Mandal
- Division of Cancer Biology, BRIC-Institute of Life Sciences, Bhubaneswar, India.
| |
Collapse
|
12
|
Colunga-Pedraza PR, Vaquera-Alfaro HA, Guzmán-Martínez Z, Alemán-Jiménez MC, Vega-Mateos A, Gómez-De León A, Villela LM, Gómez-Almaguer D. Optimizing resources: low-dose nivolumab combinations in the management of relapsed/refractory Hodgkin lymphoma. Ann Hematol 2024:10.1007/s00277-024-06098-9. [PMID: 39562359 DOI: 10.1007/s00277-024-06098-9] [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: 05/07/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024]
Abstract
Up to one-third of patients with classical Hodgkin lymphoma (cHL) are not responsive to first-line therapy or eventually relapse. Immune checkpoint inhibitors (ICIs) have been successfully employed to treat relapsed/refractory cHL (r/r cHL) but place patients at risk of financial toxicity. Early-phase trials and observational data suggest that low doses of ICIs may achieve similar results to those obtained with high doses. In this study, we report a single-center experience using low-dose nivolumab (LD-Nivo) in different combinations for r/r cHL, including monotherapy, LD-Nivo plus brentuximab vedotin (BV), and LD-Nivo plus chemotherapy. The primary outcome was to assess the efficacy of LD-nivo in patients with r/r cHL. We included 23 consecutive patients (median age 27 years; 57% female). LD-Nivo was prescribed in 40, 100, and 140 mg fixed doses Q2W. Survival analysis was performed employing the Kaplan-Meier method. 73% of patients achieved an overall response, 43% complete response, and 30% partial response. One-year overall survival was 94.4% (95% CI, 0.84-1), and the 1-year progression-free survival was 89.4% (95% CI, 0.77-1). OS and PFS were similar accross combinations. The median dose of nivolumab was 0.78 mg/kg (range, 0.62-1.11), and the median number of cycles until a response was documented was 6 (range, 2-9). During follow-up, 18 patients received transplantation (11 autologous, 6 allogeneic). No statistically significant differences in survival or response were detected between nivolumab combinations or doses. Adverse events were observed in 61% of the patients, with none grade 3-4. LD-Nivo demonstrated promising results in relapsed/refractory HL, highlighting its potential as a cost-effective treatment option. Further research is needed to validate these findings and guide clinical practice.
Collapse
Affiliation(s)
- Perla R Colunga-Pedraza
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Héctor A Vaquera-Alfaro
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Zulia Guzmán-Martínez
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Marion Carolina Alemán-Jiménez
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Antonio Vega-Mateos
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Andrés Gómez-De León
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Luis Mario Villela
- Universidad Autónoma de Sinaloa, Escuela de Medicina; Hospital General Fernando Ocaranza ISSSTE, Centro Médico Dr. Ignacio Chávez ISSSTESON, Hermosillo, Sonora, Mexico
- Unidad de Atención a La Salud, IMSS Bienestar, Ciudad de Mexico, México
- Hospital CIMA, Hospital CIMA, Hermosillo Sonora, Mexico
| | - David Gómez-Almaguer
- Servicio de Hematología, Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico.
| |
Collapse
|
13
|
Vredevoogd DW, Apriamashvili G, Levy PL, Sinha S, Huinen ZR, Visser NL, de Bruijn B, Boshuizen J, van Hal-van Veen SE, Ligtenberg MA, Bleijerveld OB, Lin CP, Díaz-Gómez J, Sánchez SD, Markovits E, Simon Nieto J, van Vliet A, Krijgsman O, Markel G, Besser MJ, Altelaar M, Ruppin E, Peeper DS. TMED inhibition suppresses cell surface PD-1 expression and overcomes T cell dysfunction. J Immunother Cancer 2024; 12:e010145. [PMID: 39510795 PMCID: PMC11552591 DOI: 10.1136/jitc-2024-010145] [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] [Accepted: 08/09/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Blockade of the programmed cell death protein 1 (PD-1) immune checkpoint (ICB) is revolutionizing cancer therapy, but little is known about the mechanisms governing its expression on CD8 T cells. Because PD-1 is induced during activation of T cells, we set out to uncover regulators whose inhibition suppresses PD-1 abundance without adversely impacting on T cell activation. METHODS To identify PD-1 regulators in an unbiased fashion, we performed a whole-genome, fluorescence-activated cell sorting (FACS)-based CRISPR-Cas9 screen in primary murine CD8 T cells. A dual-readout design using the activation marker CD137 allowed us to uncouple genes involved in PD-1 regulation from those governing general T cell activation. RESULTS We found that the inactivation of one of several members of the TMED/EMP24/GP25L/p24 family of transport proteins, most prominently TMED10, reduced PD-1 cell surface abundance, thereby augmenting T cell activity. Another client protein was cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which was also suppressed by TMED inactivation. Treatment with TMED inhibitor AGN192403 led to lysosomal degradation of the TMED-PD-1 complex and reduced PD-1 abundance in tumor-infiltrating CD8 T cells (TIL) in mice, thus reversing T cell dysfunction. Clinically corroborating these findings, single-cell RNA analyses revealed a positive correlation between TMED expression in CD8 TIL, and both a T cell dysfunction signature and lack of ICB response. Similarly, patients receiving a TIL product with high TMED expression had a shorter overall survival. CONCLUSION Our results uncover a novel mechanism of PD-1 regulation, and identify a pharmacologically tractable target whose inhibition suppresses PD-1 abundance and T cell dysfunction.
Collapse
Affiliation(s)
- David W Vredevoogd
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Georgi Apriamashvili
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Pierre L Levy
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Sanju Sinha
- Cancer Data Science Laboratory, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| | - Zowi R Huinen
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Nils L Visser
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Beaunelle de Bruijn
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Julia Boshuizen
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Susan E van Hal-van Veen
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Maarten A Ligtenberg
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Onno B Bleijerveld
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Chun-Pu Lin
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Judit Díaz-Gómez
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Santiago Duro Sánchez
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Ettai Markovits
- Ella Lemelbaum Institute for Immuno-oncology, Sheba Medical Center, Tel Hashomer, Israel
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Juan Simon Nieto
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Alex van Vliet
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Oscar Krijgsman
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Gal Markel
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Davidoff Center and Samueli Integrative Cancer Pioneering Center, Rabin Medical Center, Petah Tikva, Israel
| | - Michal J Besser
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Davidoff Center and Samueli Integrative Cancer Pioneering Center, Rabin Medical Center, Petah Tikva, Israel
- Felsenstein Medical Research Center, The Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maarten Altelaar
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| | - Daniel S Peeper
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Gandhi KA, Shirsat A, Hj SK, Chavan A, Dicholkar P, Shah S, Menon N, Noronha V, Joshi A, Prabhash K, Patil V, Gota V. Pharmacokinetics and clinical outcomes of low-dose nivolumab relative to conventional dose in patients with advanced cancer. Cancer Chemother Pharmacol 2024; 94:659-668. [PMID: 39060628 PMCID: PMC11470857 DOI: 10.1007/s00280-024-04697-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: 12/29/2023] [Accepted: 07/05/2024] [Indexed: 07/28/2024]
Abstract
PURPOSE Nivolumab is approved at various doses, including 3 mg/kg, 240 mg and 480 mg flat doses at various dosing intervals. The concept of low-dose immunotherapy is gaining traction in recent years. However, there is a need to better understand the pharmacokinetics and clinical outcomes at lower doses. METHODS Patients were either administered 40 mg flat dose or 3 mg/kg Q2W/Q3W, depending on affordability as per prevailing hospital practice. All patients were hospitalized on day 1 and pharmacokinetic samples were collected at 0, 0.5, 1.0, 6.0, 24.0, 72.0 h and day 14 following administration of the first dose of nivolumab. Plasma nivolumab levels were measured by ELISA. Patients were followed up for response and toxicity. RESULTS Twenty five patients were included in the study. Fourteen received nivolumab at conventional dose (3 mg/kg), while 11 patients received low-dose (40 mg flat). The geometric means of dose normalized Cmax and AUC0-t were comparable between those who received conventional dose and low-dose of nivolumab (0.28 versus 0.23 µg/mL/mg and 0.0014 versus 0.0011 d/mL respectively). Nineteen patients were evaluable for response. ORR among patients who received conventional dose was 5/11 (45.5%) whereas it was 4/9 (44.4%) in the low-dose cohort. All 14 (100%) patients in conventional dosing group and 7/11 patients (63.64%) in low-dose group had treatment emergent adverse events. Grade ≥ 3 toxicities were observed in 4/14 patients in conventional dose group and none in low-dose group. CONCLUSION Low-dose nivolumab leads to lower exposure in patients as compared with conventional dose, but low-dose was better tolerated, while response rates were comparable to conventional dose.
Collapse
Affiliation(s)
- Khushboo A Gandhi
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India
| | - Aditi Shirsat
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India
| | - Sharat Kumar Hj
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India
| | - Ashish Chavan
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India
| | - Parnika Dicholkar
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India
| | - Saniya Shah
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India
| | - Nandini Menon
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
- Homi bhabha National Institute, Mumbai, 400094, India
| | - Vanita Noronha
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
- Homi bhabha National Institute, Mumbai, 400094, India
| | - Amit Joshi
- Homi bhabha National Institute, Mumbai, 400094, India
- Department of Medical Oncology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, 410210, India
| | - Kumar Prabhash
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
- Homi bhabha National Institute, Mumbai, 400094, India
| | - Vijay Patil
- Department of Medical Oncology, Tata Memorial Hospital, Parel, Mumbai, 400012, India
| | - Vikram Gota
- Department of Clinical Pharmacology, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC) Sector- 22, Kharghar, Navi Mumbai, 410210, India.
- Homi bhabha National Institute, Mumbai, 400094, India.
| |
Collapse
|
15
|
Kicken MP, Deenen MJ, Moes DJAR, Hendrikx JJMA, van den Borne BEEM, Dumoulin DW, van der Wekken AJ, van den Heuvel MM, Ter Heine R. An Evidence-Based Rationale for Dose De-escalation of Subcutaneous Atezolizumab. Target Oncol 2024; 19:779-787. [PMID: 39085452 PMCID: PMC11393195 DOI: 10.1007/s11523-024-01087-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Atezolizumab is a programmed death-ligand 1 (PD-L1) checkpoint inhibitor for the treatment of different forms of cancer. The subcutaneous formulation of atezolizumab has recently received approval. However, treatment with atezolizumab continues to be expensive, and the number of patients needing treatment with this drug continues to increase. OBJECTIVE We propose two alternative dosing regimens for subcutaneous atezolizumab to reduce drug expenses while ensuring effective exposure; one may be directly implemented in the clinic. PATIENTS AND METHODS We developed two alternative dose interval prolongation strategies based on pharmacokinetic modeling and simulation. The first dosing regimen was based on patients' weight while maintaining equivalent systemic drug exposure by adhering to Food and Drug Administration (FDA) guidelines for in silico dose adjustments. The second dosing regimen aimed to have a minimum atezolizumab concentration above the 6 µg/mL threshold, associated with 95% intratumoral PD-L1 receptor saturation for at least 95% of all patients. RESULTS We found that, for the weight-based dosing regimen, the approved 3-week dosing interval could be extended to 5 weeks for patients < 50 kg and 4 weeks for patients weighing 50-65 kg. Besides improving patient convenience, these alternative dosing intervals led to a predicted 7% and 12% cost reduction for either the USA or European population. For the second dosing regimen, we predicted that a 6-week dosing interval would result in 95% of the patients above the 6 µg/mL threshold while reducing costs by 50%. CONCLUSIONS We have developed and evaluated two alternative dosing regimens that resulted in a cost reduction. Our weight-based dosing regimen can be directly implemented and complies with FDA guidelines for alternative dosing regimens of PD-L1 inhibitors. For the more progressive alternative dosing regimen aimed at the intratumoral PD-L1 receptor threshold, further evidence on efficacy and safety is needed before implementation.
Collapse
Affiliation(s)
- Mart P Kicken
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands.
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, Eindhoven, The Netherlands.
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Dirk J A R Moes
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute (NKI-AVL), Amsterdam, The Netherlands
| | | | - Daphne W Dumoulin
- Department of Pulmonary Medicine, Erasmus Medical Center Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Anthonie J van der Wekken
- Department of Pulmonology, University of Gronigen, University Medical Centre Groningen, Groningen, The Netherlands
| | | | - Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
16
|
Deng Q, Zhu L, Weiss B, Aanur P, Gao L. Strategies for successful dose optimization in oncology drug development: a practical guide. J Biopharm Stat 2024:1-15. [PMID: 39127994 DOI: 10.1080/10543406.2024.2387364] [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: 09/28/2023] [Accepted: 07/27/2024] [Indexed: 08/12/2024]
Abstract
Dose optimization is a critical challenge in drug development. Historically, dose determination in oncology has followed a divergent path from other non-oncology therapeutic areas due to the unique characteristics and requirements in Oncology. However, with the emergence of new drug modalities and mechanisms of drugs in oncology, such as immune therapies, radiopharmaceuticals, targeted therapies, cytostatic agents, and others, the dose-response relationship for efficacy and toxicity could be vastly varied compared to the cytotoxic chemotherapies. The doses below the MTD may demonstrate similar efficacy to the MTD with an improved tolerability profile, resembling what is commonly observed in non-oncology treatments. Hence, alternate strategies for dose optimization are required for new modalities in oncology drug development. This paper delves into the historical evolution of dose finding methods from non-oncology to oncology, highlighting examples and summarizing the underlying drivers of change. Subsequently, a practical framework and guidance are provided to illustrate how dose optimization can be incorporated into various stages of the development program. We provide the following general recommendations: 1) The objective for phase I is to identify a dose range rather than a single MTD dose for subsequent development to better characterize the safety and tolerability profile within the dose range. 2) At least two doses separable by PK are recommended for dose optimization in phase II. 3) Ideally, dose optimization should be performed before launching the confirmatory study. Nevertheless, innovative designs such as seamless II/III design can be implemented for dose selection and may accelerate the drug development program.
Collapse
Affiliation(s)
- Qiqi Deng
- Biostatistics and Programming, Moderna Inc., Cambridge, MA, USA
| | - Lili Zhu
- Biostatistics and Programming, Moderna Inc., Cambridge, MA, USA
| | - Brendan Weiss
- Clinical Development Oncology, Moderna Inc., Cambridge, MA, USA
| | - Praveen Aanur
- Clinical Development Oncology, Moderna Inc., Cambridge, MA, USA
| | - Lei Gao
- Biostatistics and Programming, Moderna Inc., Cambridge, MA, USA
| |
Collapse
|
17
|
Trikha M, Sarkar L, Dhanawat A, Syed N, Gujarathi H, Vora M, Sivakumar Raja AS, Bhargava P, Ramaswamy A, Mandavkar S, Saklani A, Kaushal RK, Bal M, Shetty O, Yadav S, Ostwal V. Performance of Low-Dose Immunotherapy and Standard-Dose Immunotherapy in Microsatellite Instability-High Metastatic Colorectal Cancer: Real-World Data (CLouD-High Study). JCO Glob Oncol 2024; 10:e2400141. [PMID: 39159410 DOI: 10.1200/go.24.00141] [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: 04/01/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 08/21/2024] Open
Abstract
PURPOSE Standard-dose immune checkpoint inhibitors (SD-ICIs) are the standard of care as initial therapy in microsatellite instable-high (MSI-H) advanced/metastatic colorectal adenocarcinomas (mCRC), but there are preclinical data to suggest that low-dose ICIs (LD-ICI) might also have similar efficacy. MATERIALS AND METHODS A retrospective study of patients with MSI-H mCRC receiving ICIs between June 2017 and January 2023 was conducted. The primary end point of the study was 12-month progression-free survival (PFS), which was computed using the Kaplan-Meier method. RESULTS A total of 65 patients were available for analysis during the study period. Sixty patients (92%) received nivolumab, whereas the remaining received pembrolizumab. First-line ICIs were received by 18 patients (28%), whereas 47 patients (72%) received ICIs during later lines. Thirty patients (47%) received LD-ICIs (all received nivolumab), with the remaining receiving SD-ICIs (53%). At a median follow-up of 16.5 (95% CI, 11.8 to 21.2) months, median PFS was not reached in the entire cohort. The 12-month PFS rate in the LD-ICI cohort was 90%, whereas it was 75.8% in the SD-ICI cohort. There were no statistical differences in patients receiving ICIs as first-line therapy (12 months PFS-94.4%) or during later lines of therapy (12-month PFS-77.9%; P = .56). CONCLUSION ICIs in the current study show survivals which are similar to those seen in seminal trials in patients with MSI-H mCRC. Low-dose ICIs appear to work in MSI-H mCRC and should be explored prospectively in clinical trials. Patients with MSI-H status should be exposed to ICIs, whether initially or later during treatment, whenever feasible.
Collapse
Affiliation(s)
- Mehak Trikha
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Laboni Sarkar
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Aditya Dhanawat
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Noorzia Syed
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Himanshu Gujarathi
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Manan Vora
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - A Sree Sivakumar Raja
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Sarika Mandavkar
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| | - Avanish Saklani
- Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute HBNI, Mumbai, India
| | - Rajiv Kumar Kaushal
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute HBNI, Mumbai, India
| | - Munita Bal
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute HBNI, Mumbai, India
| | - Omshree Shetty
- Department of Molecular Pathology, Tata Memorial Centre, Homi Bhabha National Institute HBNI, Mumbai, India
| | - Subhash Yadav
- Department of Pathology, Tata Memorial Centre, Homi Bhabha National Institute HBNI, Mumbai, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Parel, Mumbai, India
| |
Collapse
|
18
|
Elijah J, Puzanov I, Cresanti B, Hamad L, Attwood K, Catalfamo K, Riebandt G. Evaluation of safety outcomes between nivolumab regimens with differing dosing patterns. J Oncol Pharm Pract 2024:10781552241264817. [PMID: 39043219 DOI: 10.1177/10781552241264817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
BACKGROUND Real-world safety outcomes between the two flat-dose nivolumab regimens demonstrated to be similar in a study of adjuvant nivolumab recipients for melanoma. However, this study was limited by a single oncology patient population, a small sample size, and insufficient study power. The primary objective of this study was to evaluate the incidence of immunotherapy-related adverse effects (irAEs) between nivolumab regimens with differing dosing patterns in various solid tumor patient populations. METHODS Single-center retrospective cohort study of adult patients with solid tumor malignancies who received nivolumab 240 mg Q2W or 480 mg Q4W, or who were transitioned from 240 mg Q2W to 480 mg Q4W from March 1, 2018 to March 31, 2022 were selected for analysis from an electronic health record generated report. The primary endpoint evaluated was the incidence of irAEs. Secondary endpoints included the incidence of significant irAEs and reasons for treatment discontinuation. These endpoints were compared by univariate analysis between all three cohorts. A multivariate analysis was then conducted for the primary endpoint. RESULTS Nivolumab 240 mg Q2W was associated with a statistically significant increase in the incidence of colitis whereas the 480 mg Q4W regimen was associated with a statistically significant increase in the incidence of pruritis. The incidence of irAEs was not different between the three cohorts, while the incidence of significant irAEs was higher in the 240 mg Q2W and 240 mg Q2W to 480 mg Q4W cohorts. CONCLUSION Clinicians ought to be aware of differences in the irAE profiles between nivolumab regimens with differing dosing patterns.
Collapse
Affiliation(s)
- Joseph Elijah
- School of Pharmacy and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Igor Puzanov
- Department of Medical Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | - Lamya Hamad
- Department of Clinical Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Kayla Catalfamo
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Grazyna Riebandt
- Department of Clinical Pharmacy, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
19
|
Pin L, Villar SS, Dehbi HM. Implementing and assessing Bayesian response-adaptive randomisation for backfilling in dose-finding trials. Contemp Clin Trials 2024; 142:107567. [PMID: 38729298 DOI: 10.1016/j.cct.2024.107567] [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: 12/11/2023] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 05/12/2024]
Abstract
Traditional approaches in dose-finding trials, such as the continual reassessment method, focus on identifying the maximum tolerated dose. In contemporary early-phase dose-finding trials, especially in oncology with targeted agents or immunotherapy, a more relevant aim is to identify the lowest dose level that maximises efficacy whilst remaining tolerable. Backfilling, defined as the practice of assigning patients to dose levels lower than the current highest tolerated dose, has been proposed to gather additional pharmacokinetic, pharmacodynamic and biomarker data to recommend the most appropriate dose to carry forward for subsequent studies. The first formal framework [5] for backfilling proposed randomising backfill patients with equal probability among those doses below the dose level where the study is currently at. Here, we propose to use Bayesian response-adaptive randomisation to backfill patients. This patient-oriented approach to backfilling aims to allocate more patients to dose levels in the backfill set with higher expected efficacy based on emerging data. The backfill set constitutes of the doses below the dose the dose-finding algorithm is at. At study completion, collective patient data inform the dose-response curve, suggesting an optimal dose level balancing toxicity and efficacy. Our simulation study across diverse clinical trial settings demonstrates that a backfilling strategy using Bayesian response-adaptive randomisation can result in a patient-oriented patient assignment procedure which simultaneously enhances the likelihood of correctly identifying the most appropriate dose level. This contribution offers a methodological framework and practical implementation for patient-oriented backfilling, encompassing design and analysis considerations in early-phase trials.
Collapse
Affiliation(s)
- Lukas Pin
- MRC Biostatistics Unit at University of Cambridge, Cambridge, UK.
| | - Sofía S Villar
- MRC Biostatistics Unit at University of Cambridge, Cambridge, UK
| | - Hakim-Moulay Dehbi
- Comprehensive Clinical Trials Unit at University College London, London, UK
| |
Collapse
|
20
|
Yuan Y, Zhou H, Liu S. Statistical and practical considerations in planning and conduct of dose-optimization trials. Clin Trials 2024; 21:273-286. [PMID: 38243399 PMCID: PMC11134987 DOI: 10.1177/17407745231207085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
The U.S. Food and Drug Administration launched Project Optimus with the aim of shifting the paradigm of dose-finding and selection toward identifying the optimal biological dose that offers the best balance between benefit and risk, rather than the maximum tolerated dose. However, achieving dose optimization is a challenging task that involves a variety of factors and is considerably more complicated than identifying the maximum tolerated dose, both in terms of design and implementation. This article provides a comprehensive review of various design strategies for dose-optimization trials, including phase 1/2 and 2/3 designs, and highlights their respective advantages and disadvantages. In addition, practical considerations for selecting an appropriate design and planning and executing the trial are discussed. The article also presents freely available software tools that can be utilized for designing and implementing dose-optimization trials. The approaches and their implementation are illustrated through real-world examples.
Collapse
Affiliation(s)
- Ying Yuan
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heng Zhou
- Biostatistics and Research Decision Sciences, Merck and Co., Inc, Rahway, NJ, USA
| | - Suyu Liu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
21
|
Puszkiel A, Bianconi G, Pasquiers B, Balakirouchenane D, Arrondeau J, Boudou-Rouquette P, Bretagne MC, Salem JE, Declèves X, Vidal M, Kramkimel N, Guegan S, Aractingi S, Huillard O, Alexandre J, Wislez M, Goldwasser F, Blanchet B. Extending the dosing intervals of nivolumab: model-based simulations in unselected cancer patients. Br J Cancer 2024; 130:1866-1874. [PMID: 38532102 PMCID: PMC11130267 DOI: 10.1038/s41416-024-02659-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: 10/03/2023] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Reducing nivolumab dose intensity could increase patients' life quality and decrease the financial burden while maintaining efficacy. The aims of this study were to develop a population PK model of nivolumab based on data from unselected metastatic cancer patients and to simulate extended-interval regimens allowing to maintain minimal effective plasma concentrations (MEPC). METHODS Concentration-time data (992 plasma nivolumab concentrations, 364 patients) were modeled using a two-compartment model with linear elimination clearance in Monolix software. Extended-interval regimens allowing to maintain steady-state trough concentrations (Cmin,ss) above the MEPC of 2.5 mg/L or 1.5 mg/L in >90% of patients were simulated. RESULTS Increasing 3-times the dosing interval from 240 mg every two weeks (Q2W) to Q6W and 2-times from 480 mg Q4W to Q8W resulted in Cmin,ss above 2.5 mg/L in 95.8% and 95.4% of patients, respectively. 240 mg Q8W and 480 mg Q10W resulted in Cmin,ss above 1.5 mg/L in 91.0% and 91.8% of patients, respectively. Selection of a 240 mg Q6W regimen would decrease by 3-fold the annual treatment costs compared to standard regimen of 240 mg Q2W (from 78,744€ to 26,248€ in France). CONCLUSIONS Clinical trials are warranted to confirm the non-inferiority of extended-interval compared to standard regimen.
Collapse
Affiliation(s)
- Alicja Puszkiel
- Université Paris Cité, Faculté de Pharmacie de Paris, INSERM UMR-S1144, Paris, France.
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France.
| | - Guillaume Bianconi
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
| | - Blaise Pasquiers
- Université Paris Cité, Faculté de Pharmacie de Paris, INSERM UMR-S1144, Paris, France
- PhinC Development, Massy, France
| | | | - Jennifer Arrondeau
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
| | - Pascaline Boudou-Rouquette
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
| | - Marie-Claire Bretagne
- Department of Pharmacology, Pharmacovigilance Unit, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
| | - Joe-Elie Salem
- Department of Pharmacology, Pharmacovigilance Unit, Pitié-Salpêtrière Hospital, AP-HP, Paris, France
- INSERM, CIC-1901, Sorbonne Université, Paris, France
| | - Xavier Declèves
- Université Paris Cité, Faculté de Pharmacie de Paris, INSERM UMR-S1144, Paris, France
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
| | - Michel Vidal
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
- Université Paris Cité, Faculté de Pharmacie de Paris, UMR8038 CNRS CiTCoM, U1268 INSERM, CARPEM, Paris, France
| | - Nora Kramkimel
- Department of Dermatology, Cochin University Hospital, AP-HP, Paris, France
| | - Sarah Guegan
- Department of Dermatology, Cochin University Hospital, AP-HP, Paris, France
| | - Selim Aractingi
- Department of Dermatology, Cochin University Hospital, AP-HP, Paris, France
| | - Olivier Huillard
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
| | - Jérôme Alexandre
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
- Université Paris Cité, INSERM, Centre de Recherche des Cordeliers, Équipe labélisée Ligue Contre le Cancer, CNRS SNC 5096, Sorbonne Université, Paris, France
| | - Marie Wislez
- Department of Pneumology, Cochin University Hospital, AP-HP, Paris, France
| | - François Goldwasser
- Department of Medical Oncology, Cochin University Hospital, Institut du Cancer Paris CARPEM, AP-HP, Paris, France
- Université Paris Cité, Faculté de Médecine, INSERM, U1016, Institut Cochin, Paris, France
| | - Benoit Blanchet
- Biologie du Médicament - Toxicologie, Cochin University Hospital, AP-HP, Paris, France
- Université Paris Cité, Faculté de Pharmacie de Paris, UMR8038 CNRS CiTCoM, U1268 INSERM, CARPEM, Paris, France
| |
Collapse
|
22
|
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
|
23
|
Ikeda G, Miyakoshi J, Yamamoto S, Kato K. Nivolumab in unresectable advanced, recurrent or metastatic esophageal squamous cell carcinoma. Future Oncol 2024; 20:665-677. [PMID: 38126175 DOI: 10.2217/fon-2022-1092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Esophageal cancer (EC) is the eighth most common cancer worldwide. In view of biology and anatomical restrictions, multimodality treatment strategies have been developed for EC. However, the prognosis of patients with advanced EC remains especially poor. Immunotherapy, such as PD-1/PD-L1 and CTLA-4/B7 blockade, has emerged as a potent treatment for many types of cancer and has been approved in many countries. Based on the results of the ATTRACTION-3 trial, nivolumab, an anti-PD-1 monoclonal antibody, was approved by the US FDA for patients with platinum-resistant, unresectable, recurrent or metastatic esophageal squamous cell carcinoma. The CheckMate 648 trial demonstrated that the combination of nivolumab with platinum-based fluoropyrimidine chemotherapy and combination immunotherapy with nivolumab and ipilimumab, an anti-CTLA-4 monoclonal antibody, showed a survival benefit in patients with advanced esophageal squamous cell carcinoma compared with doublet chemotherapy. This review focuses on nivolumab-containing treatments for patients with advanced esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Go Ikeda
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Gastroenterology, Nippon Medical School Graduate School of Medicine, 1-1-5, Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan
| | - Jun Miyakoshi
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shun Yamamoto
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ken Kato
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
24
|
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
|
25
|
Hu Z, Liu S, Zhao Y, Du S, Hamuro L, Shen J, Roy A, Zhu L. Nivolumab and ipilimumab population pharmacokinetics in support of pediatric dose recommendations-Going beyond the body-size effect. CPT Pharmacometrics Syst Pharmacol 2024; 13:476-493. [PMID: 38115545 PMCID: PMC10941504 DOI: 10.1002/psp4.13098] [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: 05/17/2023] [Revised: 10/22/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Body size has historically been considered the primary source of difference in the pharmacokinetics (PKs) of monoclonal antibodies (mAbs) between children aged greater than or equal to 2 years and adults. The contribution of age-associated differences (e.g., ontogeny) beyond body-size differences in the pediatric PKs of mAbs has not been comprehensively evaluated. In this study, the population PK of two mAbs (nivolumab and ipilimumab) in pediatric oncology patients were characterized. The effects of age-related covariates on nivolumab or ipilimumab PKs were assessed using data from 13 and 10 clinical studies, respectively, across multiple tumor types, including melanoma, lymphoma, central nervous system tumors (CNSTs), and other solid tumors. Clearance was lower in pediatric patients (aged 1-17 years) with solid tumors or CNST than in adults after adjusting for other covariates, including the effect of body size. In contrast, clearance was similar in pediatric patients with lymphoma to that in adults with lymphoma. The pediatric effects characterized have increased the accuracy of the predictions of the model, facilitating its use in subsequent exposure comparisons between pediatric and adult patients, as well as for exposure-response analyses to inform pediatric dosing. This study approach may be applicable to the optimization of pediatric dosing of other mAbs and possibly other biologics.
Collapse
Affiliation(s)
- Zheyi Hu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Sihang Liu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Yue Zhao
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | | | | - Jun Shen
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Li Zhu
- Bristol Myers SquibbPrincetonNew JerseyUSA
| |
Collapse
|
26
|
Yu JE, Yeo IJ, Han SB, Yun J, Kim B, Yong YJ, Lim YS, Kim TH, Son DJ, Hong JT. Significance of chitinase-3-like protein 1 in the pathogenesis of inflammatory diseases and cancer. Exp Mol Med 2024; 56:1-18. [PMID: 38177294 PMCID: PMC10834487 DOI: 10.1038/s12276-023-01131-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 08/06/2023] [Accepted: 08/28/2023] [Indexed: 01/06/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that mediates inflammation, macrophage polarization, apoptosis, and carcinogenesis. The expression of CHI3L1 is strongly upregulated by various inflammatory and immunological diseases, including several cancers, Alzheimer's disease, and atherosclerosis. Several studies have shown that CHI3L1 can be considered as a marker of disease diagnosis, prognosis, disease activity, and severity. In addition, the proinflammatory action of CHI3L1 may be mediated via responses to various proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interferon-γ. Therefore, CHI3L1 may contribute to a vast array of inflammatory diseases. However, its pathophysiological and pharmacological roles in the development of inflammatory diseases remain unclear. In this article, we review recent findings regarding the roles of CHI3L1 in the development of inflammatory diseases and suggest therapeutic approaches that target CHI3L1.
Collapse
Affiliation(s)
- Ji Eun Yu
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
- College of Pharmacy, Kyungpook National University, 80 Daehakro, Bukgu, Daegu, 41566, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Bongcheol Kim
- Senelix Co. Ltd., 25, Beobwon-ro 11-gil, Songpa-gu, Seoul, 05836, Republic of Korea
| | - Yoon Ji Yong
- PRESTI GEBIOLOGICS Co. Ltd., Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28161, Republic of Korea
| | - Young-Soo Lim
- PRESTI GEBIOLOGICS Co. Ltd., Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28161, Republic of Korea
| | - Tae Hun Kim
- Autotelic Bio Inc., Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungbuk, 28160, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, 194-31, Osongsaengmyeong 1-ro, Osong-eup, Cheongju-si, Chungbuk, 28160, Republic of Korea.
| |
Collapse
|
27
|
Proctor JR, Wong H. Time-dependent clearance can confound exposure-response analysis of therapeutic antibodies: A comprehensive review of the current literature. Clin Transl Sci 2024; 17:e13676. [PMID: 37905360 PMCID: PMC10766027 DOI: 10.1111/cts.13676] [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: 09/01/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023] Open
Abstract
Exposure-response (ER) analysis is used to optimize dose and dose regimens during clinical development. Characterization of relationships between drug exposure and efficacy or safety outcomes can be utilized to make dose adjustments that improve patient response. Therapeutic antibodies typically show predictable pharmacokinetics (PK) but can exhibit clearance that decreases over time due to treatment. Moreover, time-dependent changes in clearance are frequently associated with drug response, with larger decreases in clearance and increased exposure seen in patients who respond to treatment. This often confounds traditional ER analysis, as drug response influences exposure rather than the reverse. In this review, we survey published population PK analyses for reported time-dependent drug clearance effects across 158 therapeutic antibodies approved or in regulatory review. We describe the mechanisms by which time-dependent clearance can arise, and evaluate trends in frequency, magnitude, and time scale of changes in clearance with respect to indication, mechanistic interpretation of time-dependence, and PK modeling techniques employed. We discuss the modeling and simulation strategies commonly used to characterize time-dependent clearance, and examples where time-dependent clearance has impeded ER analysis. A case study using population model simulation was explored to interrogate the impact of time-dependent clearance on ER analysis and how it can lead to spurious conclusions. Overall, time-dependent clearance arises frequently among therapeutic antibodies and has spurred erroneous conclusions in ER analysis. Appropriate PK modeling techniques aid in identifying and characterizing temporal shifts in exposure that may impede accurate ER assessment and successful dose optimization.
Collapse
Affiliation(s)
- Jeffrey R. Proctor
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Harvey Wong
- Faculty of Pharmaceutical SciencesThe University of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
28
|
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
|
29
|
Yates JWT, Mistry HB. Skipping a pillar does not make for strong foundations: Pharmacokinetic-pharmacodynamic reasoning behind the shape of dose-response relationships in oncology. CPT Pharmacometrics Syst Pharmacol 2023; 12:1591-1601. [PMID: 37771203 PMCID: PMC10681527 DOI: 10.1002/psp4.13020] [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: 05/31/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 09/30/2023] Open
Abstract
Dose-response analysis is often applied to the quantification of drug-effect especially for slowly responding disease end points where a comparison is made across dose levels after a particular period of treatment. It has long been recognized that exposure - response is more appropriate than dose-response. However, trials necessarily are designed as dose-response experiments. Second, a wide range of functional forms are used to express relationships between dose and response. These considerations are also important for clinical development because pharmacokinetic (PK; and variability) plus pharmacokinetic-pharmacodynamic modeling may allow one to anticipate the shape of the dose-response curve and so the trial design. Here, we describe how the location and steepness of the dose response is determined by the PKs of the compound being tested and its exposure-response relationship in terms of potency (location), efficacy (maximum effect) and Hill coefficient (steepness). Thus, the location (50% effective dose [ED50 ]) is dependent not only on the potency (half-maximal effective concentration) but also the compound's PKs. Similarly, the steepness of the dose response is shown to be a function of the half-life of the drug. It is also shown that the shape of relationship varies dependent on the assumed time course of the disease. This is important in the context of drug-discovery where the in vivo potencies of compounds are compared as well as when considering an analysis of summary data (for example, model-based meta-analysis) for clinical decision making.
Collapse
|
30
|
Kumar V, Wei XX. Immune Checkpoint Inhibitors in Metastatic Bladder and Other Solid Malignancies: How Long is Enough? Bladder Cancer 2023; 9:201-210. [PMID: 38993185 PMCID: PMC11181755 DOI: 10.3233/blc-230039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/24/2023] [Indexed: 07/13/2024]
Abstract
The introduction of T-cell targeted immunomodulators blocking the PD-1 and PD-L1 axis is unquestionably one of the most notable advancements in the treatment of advanced or metastatic solid malignancies, including bladder cancer. Immune checkpoint antibodies are now widely utilized as monotherapies or in combination with other systemic therapies in the first or subsequent lines of treatment in approximately 50 cancer types. Deep and durable responses and long tails of survival curves are hallmarks of patients treated with immune checkpoint inhibitors. However, treatment can have negative impacts, including serious treatment-related side effects as well as a high financial burden to individual patients and the healthcare system. There is increasing data that the benefit of immune checkpoint treatment may persist after treatment is discontinued for reasons other than progressive disease, particularly in patients who have achieved a durable complete response. However, the optimal treatment duration and activity after treatment reinitiation remains undefined and will likely be influenced by disease biology (histology and genomics), treatment (monotherapy or combination therapy), and disease context (depth and duration of response). Well-designed prospective clinical trials and the development and validation of biomarkers that predict outcomes after treatment cessation are needed to move the field forward.
Collapse
Affiliation(s)
- Vivek Kumar
- Brigham and Women’s Hospital, Boston, MA, USA
| | - Xiao X. Wei
- Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
31
|
Porter LH, Zhu JJ, Lister NL, Harrison SG, Keerthikumar S, Goode DL, Urban RQ, Byrne DJ, Azad A, Vela I, Hofman MS, Neeson PJ, Darcy PK, Trapani JA, Taylor RA, Risbridger GP. Low-dose carboplatin modifies the tumor microenvironment to augment CAR T cell efficacy in human prostate cancer models. Nat Commun 2023; 14:5346. [PMID: 37660083 PMCID: PMC10475084 DOI: 10.1038/s41467-023-40852-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 08/11/2023] [Indexed: 09/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have transformed the treatment landscape for hematological malignancies. However, CAR T cells are less efficient against solid tumors, largely due to poor infiltration resulting from the immunosuppressive nature of the tumor microenvironment (TME). Here, we assessed the efficacy of Lewis Y antigen (LeY)-specific CAR T cells in patient-derived xenograft (PDX) models of prostate cancer. In vitro, LeY CAR T cells directly killed organoids derived from androgen receptor (AR)-positive or AR-null PDXs. In vivo, although LeY CAR T cells alone did not reduce tumor growth, a single prior dose of carboplatin reduced tumor burden. Carboplatin had a pro-inflammatory effect on the TME that facilitated early and durable CAR T cell infiltration, including an altered cancer-associated fibroblast phenotype, enhanced extracellular matrix degradation and re-oriented M1 macrophage differentiation. In a PDX less sensitive to carboplatin, CAR T cell infiltration was dampened; however, a reduction in tumor burden was still observed with increased T cell activation. These findings indicate that carboplatin improves the efficacy of CAR T cell treatment, with the extent of the response dependent on changes induced within the TME.
Collapse
Affiliation(s)
- L H Porter
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - J J Zhu
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - N L Lister
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia
| | - S G Harrison
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, 3800, Australia
| | - S Keerthikumar
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - D L Goode
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - R Quezada Urban
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - D J Byrne
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - A Azad
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - I Vela
- Queensland Bladder Cancer Initiative, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Center, School of Biomedical Science, Faculty of Health, Queensland University of Technology, Brisbane, QLD, 4102, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, 4102, Australia
| | - M S Hofman
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - P J Neeson
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - P K Darcy
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - J A Trapani
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - R A Taylor
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Physiology, Monash University, Clayton, VIC, 3800, Australia.
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
| | - G P Risbridger
- Prostate Cancer Research Group, Monash Biomedicine Discovery Institute, Cancer Program, Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, 3800, Australia.
- Cancer Immunology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
| |
Collapse
|
32
|
Sangro B, Yau T, El‐Khoueiry AB, Kudo M, Shen Y, Tschaika M, Roy A, Feng Y, Gao L, Aras U. Exposure-response analysis for nivolumab plus ipilimumab combination therapy in patients with advanced hepatocellular carcinoma (CheckMate 040). Clin Transl Sci 2023; 16:1445-1457. [PMID: 37165980 PMCID: PMC10432868 DOI: 10.1111/cts.13544] [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: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
This analysis was conducted to inform dose selection of a combination of nivolumab plus ipilimumab for the treatment of sorafenib-experienced patients with hepatocellular carcinoma (HCC). CheckMate 040 is an open-label, multicohort, phase I/II trial in adults with advanced HCC that evaluated nivolumab monotherapy (0.1-10 mg/kg once every 2 weeks [q2w]) and the following three combinations of nivolumab plus ipilimumab: (1) nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks (q3w) for four doses, followed by nivolumab monotherapy 240 mg q2w (arm A); (2) nivolumab 3 mg/kg plus ipilimumab 1 mg/kg q3w for four doses, followed by nivolumab monotherapy 240 mg q2w (arm B); and (3) nivolumab 3 mg/kg q2w plus ipilimumab 1 mg/kg every 6 weeks continuously (arm C). Exposure-response relationships (efficacy and safety) were characterized using nivolumab and ipilimumab concentrations after the first dose (Cavg1) as the exposure measure. Objective tumor response (OTR) and overall survival (OS) improvements were associated with increased ipilimumab exposure (OTR: odds ratio 1.45, 95% confidence interval [CI], 1.13-1.86; OS: hazard ratio 0.86, 95% CI 0.75-0.98), but not nivolumab exposure (OTR: odds ratio 0.99, 95% CI 0.97-1.02; OS: hazard ratio 1.08, 95% CI 0.89-1.32). Hepatic treatment-related and immune-mediated adverse events were more common in arm A than in arms B or C. Nivolumab 1 mg/kg plus ipilimumab 3 mg/kg q3w for four doses, followed by nivolumab monotherapy 240 mg q2w had the most favorable benefit:risk profile in patients with advanced HCC.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver UnitClinica Universidad de Navarra‐IDISNA and CIBEREHDPamplonaSpain
| | - Thomas Yau
- University of Hong Kong, Hong Kong Special Administrative RegionPokfulamChina
| | | | - Masatoshi Kudo
- Department of Gastroenterology and HepatologyKindai University Faculty of MedicineOsakaJapan
| | - Yun Shen
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Yan Feng
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Ling Gao
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Urvi Aras
- Bristol Myers SquibbPrincetonNew JerseyUSA
| |
Collapse
|
33
|
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
|
34
|
Bryant AK, Chopra Z, Edwards DM, Whalley AS, Bazzell BG, Moeller JA, Kelley MJ, Fendrick AM, Kerr EA, Ramnath N, Green MD, Hofer TP, Strohbehn GW. Adopting Weight-Based Dosing With Pharmacy-Level Stewardship Strategies Could Reduce Cancer Drug Spending By Millions. Health Aff (Millwood) 2023; 42:946-955. [PMID: 37406228 PMCID: PMC10985582 DOI: 10.1377/hlthaff.2023.00102] [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] [Indexed: 07/07/2023]
Abstract
Immune checkpoint inhibitors, a class of drugs used in approximately forty unique cancer indications, are a sizable component of the economic burden of cancer care in the US. Instead of personalized weight-based dosing, immune checkpoint inhibitors are most commonly administered at "one-size-fits-all" flat doses that are higher than necessary for the vast majority of patients. We hypothesized that personalized weight-based dosing along with common stewardship efforts at the pharmacy level, such as dose rounding and vial sharing, would lead to reductions in immune checkpoint inhibitor use and lower spending. Using data from the Veterans Health Administration (VHA) and Medicare drug prices, we estimated reductions in immune checkpoint inhibitor use and spending that would be associated with pharmacy-level stewardship strategies, in a case-control simulation study of individual patient-level immune checkpoint inhibitor administration events. We identified baseline annual VHA spending for these drugs of approximately $537 million. Combining weight-based dosing, dose rounding, and pharmacy-level vial sharing would generate expected annual VHA health system savings of $74 million (13.7 percent). We conclude that adoption of pharmacologically justified immune checkpoint inhibitor stewardship measures would generate sizable reductions in spending for these drugs. Combining these operational innovations with value-based drug price negotiation enabled by recent policy changes may improve the long-term financial viability of cancer care in the US.
Collapse
Affiliation(s)
- Alex K Bryant
- Alex K. Bryant, University of Michigan, Ann Arbor, Michigan
| | | | | | - Adam S Whalley
- Adam S. Whalley, Veterans Affairs (VA) Maine Health Care, Augusta, Maine
| | - Brian G Bazzell
- Brian G. Bazzell, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | | | - Michael J Kelley
- Michael J. Kelley, Duke University and VA National Oncology Program Office, Durham, North Carolina
| | | | - Eve A Kerr
- Eve A. Kerr, University of Michigan and VA Ann Arbor Healthcare System
| | | | | | - Timothy P Hofer
- Timothy P. Hofer, University of Michigan and VA Ann Arbor Healthcare System
| | | |
Collapse
|
35
|
Rathinasamy N, Muthu S, Krishnan A. Low-dose immunotherapy as a potentiator to increase the response with neo-adjuvant chemotherapy in oral cancers. World J Clin Cases 2023; 11:3976-3979. [PMID: 37388807 PMCID: PMC10303620 DOI: 10.12998/wjcc.v11.i17.3976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Neo-adjuvant chemotherapy (NACT) is utilized in locally advanced oral cancers to reduce the tumor burden and downstage the tumor to be amenable for definitive surgical management. Its long-term results compared to upfront surgical resection was not encouraging. Immunotherapy has now been used not only in recurrence and metastatic setting but also in the locally advanced tumor management regimens. The purpose of this concept paper is to bring forward the rationale to use a fixed low-dose immunotherapy agent as a potentiator to the standard NACT regimen and recommend their future investigation in oral cancer management.
Collapse
Affiliation(s)
- Narmadha Rathinasamy
- Department of Medical Oncology, PSG Institute of Medical Science and Research, Coimbatore 641004, Tamil Nadu, India
| | - Sathish Muthu
- Department of Orthopaedics, Orthopaedic Research Group, Coimbatore 641045, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, New Delhi 201310, Uttar Pradesh, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Anand Krishnan
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
36
|
Ter Heine R, van den Heuvel MM, Piet B, Deenen MJ, van der Wekken AJ, Hendriks LEL, Croes S, van Geel RMJM, Jansman FGA, Boshuizen RC, Franssen EJF, Smit AAJ, Dumoulin DW, Oude Munnink TH, Smit EF, Derijks HJ, van der Leest CH, Hendrikx JJMA, Moes DJAR, de Rouw N. A Systematic Evaluation of Cost-Saving Dosing Regimens for Therapeutic Antibodies and Antibody-Drug Conjugates for the Treatment of Lung Cancer. Target Oncol 2023; 18:441-450. [PMID: 37081309 DOI: 10.1007/s11523-023-00958-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 04/22/2023]
Abstract
BACKGROUND Expensive novel anticancer drugs put a serious strain on healthcare budgets, and the associated drug expenses limit access to life-saving treatments worldwide. OBJECTIVE We aimed to develop alternative dosing regimens to reduce drug expenses. METHODS We developed alternative dosing regimens for the following monoclonal antibodies used for the treatment of lung cancer: amivantamab, atezolizumab, bevacizumab, durvalumab, ipilimumab, nivolumab, pembrolizumab, and ramucirumab; and for the antibody-drug conjugate trastuzumab deruxtecan. The alternative dosing regimens were developed by means of modeling and simulation based on the population pharmacokinetic models developed by the license holders. They were based on weight bands and the administration of complete vials to limit drug wastage. The resulting dosing regimens were developed to comply with criteria used by regulatory authorities for in silico dose development. RESULTS We found that alternative dosing regimens could result in cost savings that range from 11 to 28%, and lead to equivalent pharmacokinetic exposure with no relevant increases in variability in exposure. CONCLUSIONS Dosing regimens based on weight bands and the use of complete vials to reduce drug wastage result in less expenses while maintaining equivalent exposure. The level of evidence of our proposal is the same as accepted by regulatory authorities for the approval of alternative dosing regimens of other monoclonal antibodies in oncology. The proposed alternative dosing regimens can, therefore, be directly implemented in clinical practice.
Collapse
Affiliation(s)
- Rob Ter Heine
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands.
| | | | - Berber Piet
- Department of Pulmonology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital Eindhoven, Eindhoven, The Netherlands
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Anthonie J van der Wekken
- Department of Pulmonary Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW-School for Oncology and Reproduction, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sander Croes
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Robin M J M van Geel
- Department of Clinical Pharmacy and Toxicology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Frank G A Jansman
- Department of Clinical Pharmacy, Deventer Hospital, Deventer, The Netherlands
- Unit of PharmacoTherapy, Epidemiology and Economics, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, Groningen, The Netherlands
| | | | - Eric J F Franssen
- Department of Clinical Pharmacy, Onze Lieve Vrouwe Gasthuis Hospital AC, Amsterdam, The Netherlands
| | - Arthur A J Smit
- Department of Pulmonology, Onze Lieve Vrouwe Gasthuis Hospital AC, Amsterdam, The Netherlands
| | - Daphne W Dumoulin
- Department of Pulmonary Medicine, Erasmus MC Cancer Institute, University Medical Center, Rotterdam, The Netherlands
| | - Thijs H Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Egbert F Smit
- Department of Pulmonology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Hieronymus J Derijks
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
- Department of Pharmacy, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | | | - Jeroen J M A Hendrikx
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute (NKI-AVL), Amsterdam, The Netherlands
| | - Dirk J A R Moes
- Department of Clinical Pharmacology and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Nikki de Rouw
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Geert Grooteplein Zuid 10, 6525GA, Nijmegen, The Netherlands
- Department of Clinical Pharmacy, Amphia Hospital, Breda, The Netherlands
| |
Collapse
|
37
|
Qiu Y, Zhao Y, Liu H, Cao S, Zhang C, Zang Y. Modified isotonic regression based phase I/II clinical trial design identifying optimal biological dose. Contemp Clin Trials 2023; 127:107139. [PMID: 36870476 PMCID: PMC10065963 DOI: 10.1016/j.cct.2023.107139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Conventional phase I/II clinical trial designs often use complicated parametric models to characterize the dose-response relationships and conduct the trials. However, the parametric models are hard to justify in practice, and the misspecification of parametric models can lead to substantially undesirable performances in phase I/II trials. Moreover, it is difficult for the physicians conducting phase I/II trials to clinically interpret the parameters of these complicated models, and such significant learning costs impede the translation of novel statistical designs into practical trial implementation. To solve these issues, we propose a transparent and efficient phase I/II clinical trial design, referred to as the modified isotonic regression-based design (mISO), to identify the optimal biological doses for molecularly targeted agents and immunotherapy. The mISO design makes no parametric model assumptions on the dose-response relationship and yields desirable performances under any clinically meaningful dose-response curves. The concise, clinically interpretable dose-response models and dose-finding algorithm make the proposed designs highly translational from the statistical community to the clinical community. We further extend the mISO design and develop the mISO-B design to handle the delayed outcomes. Our comprehensive simulation studies show that the mISO and mISO-B designs are highly efficient in optimal biological dose selection and patients allocation and outperform many existing phase I/II clinical trial designs. We also provide a trial example to illustrate the practical implementation of the proposed designs. The software for simulation and trial implementation are available for free download.
Collapse
Affiliation(s)
- Yingjie Qiu
- Department of Biostatistics and Health Data Science, Indiana University, USA
| | - Yi Zhao
- Department of Biostatistics and Health Data Science, Indiana University, USA
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Cancer Institute of New Jersey, Rutgers University, USA
| | - Sha Cao
- Department of Biostatistics and Health Data Science, Indiana University, USA; Center of Computational Biology and Bioinformatics, Indiana University, USA
| | - Chi Zhang
- Center of Computational Biology and Bioinformatics, Indiana University, USA; Department of Medical and Molecular Genetics, Indiana University, USA
| | - Yong Zang
- Department of Biostatistics and Health Data Science, Indiana University, USA; Center of Computational Biology and Bioinformatics, Indiana University, USA.
| |
Collapse
|
38
|
Chen C, Fan X, Zhang L, Xu P, Zou H, Zhao X, Gupta M, Feng YS, Xu XS, Yan X. Clearance as an Early Indicator of Efficacy for Therapeutic Monoclonal Antibodies: Circumventing Dose Selection Challenges in Oncology. Clin Pharmacokinet 2023; 62:705-713. [PMID: 36930421 DOI: 10.1007/s40262-023-01231-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/22/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND AND OBJECTIVE The designs of first-in-human (FIH) studies in oncology (e.g., 3 + 3 dose escalation design) usually do not provide a sufficient sample size to determine the dose-response relationship for efficacy. This study aimed to assess the feasibility of using monoclonal antibody (mAb) clearance as a biomarker for efficacy to facilitate the identification of potentially efficacious doses across cancer types and drug targets. METHODS We performed electronic searches of the Drugs@FDA website, the European Medicines Agency website, and PubMed to identify reports of FIH trials of approved mAbs in oncology. The clearance, half-life, and overall response rate (ORR) data for the mAbs at different dose levels were extracted. RESULTS Twenty-five approved mAbs were included in this study. As expected, due to the small sample sizes in FIH studies, there was no clear dose-response for ORR. However, we found a clear negative association between mAb clearance and ORR across tumors/drug targets, and a clear negative dose-clearance relationship, with clearance decreasing and saturated at high dose levels. The approved mAb doses (1-25 mg/kg) are approximately 2-fold the saturation doses (1-10 mg/kg). The associated clearance values at the approved doses vary across different cancers and drug targets (0.17-1.56 L/day), while tend to be similar within a disease/drug target. Anti-CD20 mAbs for B-cell lymphomas show a higher clearance (~ 1 L/day) than other cancers and targets (e.g., ~ 0.3 L/day for anti-PD-1). CONCLUSIONS Clearance of mAbs can be a tumor/drug target-agnostic biomarker for potential anti-tumor activity as clearance decreases with increasing ORR. Our findings shed important insights into target clearance values that may lead to desired efficacy for different cancers and drug targets, which can be used to guide dose selection for the future development of mAbs during FIH oncology studies.
Collapse
Affiliation(s)
- Chengcong Chen
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Xiaoqing Fan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Lin Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Peng Xu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Huixi Zou
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Xing Zhao
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR
| | - Manish Gupta
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA
| | - Yan Summer Feng
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA
| | - Xu Steven Xu
- Clinical Pharmacology and Quantitative Science, Genmab Inc., Princeton, NJ, USA.
| | - Xiaoyu Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR.
| |
Collapse
|
39
|
Dehbi HM, O'Quigley J, Iasonos A. Controlled amplification in oncology dose-finding trials. Contemp Clin Trials 2023; 125:107021. [PMID: 36526255 PMCID: PMC11134416 DOI: 10.1016/j.cct.2022.107021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/23/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
In oncology clinical trials the guiding principle of model-based dose-finding designs for cytotoxic agents is to progress as fast as possible towards, and identify, the dose level most likely to be the MTD. Recent developments with non-cytotoxic agents have broadened the scope of early phase trials to include multiple objectives. The ultimate goal of dose-finding designs in our modern era is to collect the relevant information in the study for final RP2D determination. While some information is collected on dose levels below and in the vicinity of the MTD during the escalation (using conventional tools such as the Continual Reassessment Method for example), designs that include expansion cohorts or backfill patients effectively amplify the information collected on the lower dose levels. This is achieved by allocating patients to dose levels slightly differently during the study in order to take into account the possibility that "less (dose) might be more". The objective of this paper is to study the concept of amplification. Under the heading of controlled amplification we can include dose expansion cohorts and backfill patients among others. We make some general observations by defining these concepts more precisely and study a specific design that exploits the concept of controlled amplification.
Collapse
Affiliation(s)
| | - John O'Quigley
- Department of Statistical Science, University College London, London, UK
| | | |
Collapse
|
40
|
Mitchell AP, Goldstein DA. Cost Savings and Increased Access With Ultra-Low-Dose Immunotherapy. J Clin Oncol 2023; 41:170-172. [PMID: 36265102 PMCID: PMC9839306 DOI: 10.1200/jco.22.01711] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/03/2022] [Accepted: 08/12/2022] [Indexed: 01/19/2023] Open
Affiliation(s)
- Aaron P. Mitchell
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel A. Goldstein
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
- Clalit Health Service, Tel Aviv-Yafo, Israel
- Optimal Cancer Care Alliance, Ann Arbor, MI
| |
Collapse
|
41
|
Shchelokov D, Demin Jr O. Receptor occupancy assessment and interpretation in terms of quantitative systems pharmacology: nivolumab case study. MAbs 2023; 15:2156317. [PMID: 36524835 PMCID: PMC9762804 DOI: 10.1080/19420862.2022.2156317] [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] [Indexed: 12/23/2022] Open
Abstract
Receptor occupancy assays applied in clinical studies provide insights into pharmacokinetic-pharmacodynamic relationships for therapeutic antibodies. When measured by different assays, however, receptor occupancy results can be controversial, as was observed for nivolumab, a monoclonal antibody targeting programmed cell death 1 (PD-1) receptor. We suggested an explanation of results obtained and a mechanistic approach based on specific features of the receptor occupancy assays: measurement of the free or bound receptor, normalized to the baseline or at each time point. The approach was evaluated against controversial clinical data on PD-1 receptor occupancy by nivolumab. It was shown that receptor occupancy measured by different assays might vary substantially if the internalization rate of the bound receptor is higher than the rate of degradation of the free receptor. Equations proposed in this work can be applied in quantitative systems pharmacology models to describe target receptor occupancy by different therapeutic antibodies.
Collapse
Affiliation(s)
- Dmitry Shchelokov
- InSysBio UK Limited, Edinburgh, UK,CONTACT Dmitry Shchelokov InSysBio UK Limited, 17-19 East London Street, EdinburghEH7 4ZD, UK
| | | |
Collapse
|
42
|
Yoshimoto S, Chester N, Xiong A, Radaelli E, Wang H, Brillantes M, Gulendran G, Glassman P, Siegel DL, Mason NJ. Development and pharmacokinetic assessment of a fully canine anti-PD-1 monoclonal antibody for comparative translational research in dogs with spontaneous tumors. MAbs 2023; 15:2287250. [PMID: 38047502 PMCID: PMC10793675 DOI: 10.1080/19420862.2023.2287250] [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: 07/11/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023] Open
Abstract
PD-1 checkpoint inhibitors have revolutionized the treatment of patients with different cancer histologies including melanoma, renal cell carcinoma, and non-small cell lung carcinoma. However, only a subset of patients show a dramatic clinical response to treatment. Despite intense biomarker discovery efforts, no single robust, prognostic correlation has emerged as a valid outcome predictor. Immune competent, pet dogs develop spontaneous tumors that share similar features to human cancers including chromosome aberrations, molecular subtypes, immune signatures, tumor heterogeneity, metastatic behavior, and chemotherapeutic response. As such, they represent a valuable parallel patient population in which to investigate predictive biomarkers of checkpoint inhibition. However, the lack of a validated, non-immunogenic, canine anti-PD-1 antibody for pre-clinical use hinders this comparative approach and prevents potential clinical benefits of PD-1 blockade being realized in the veterinary clinic. To address this, fully canine single-chain variable fragments (scFvs) that bind canine (c)PD-1 were isolated from a comprehensive canine scFv phage display library. Lead candidates were identified that bound with high affinity to cPD-1 and inhibited its interaction with canine PD-L1 (cPD-L1). The lead scFv candidate re-formatted into a fully canine IgGD reversed the inhibitory effects of cPD-1:cPD-L1 interaction on canine chimeric antigen receptor (CAR) T cell function. In vivo administration showed no toxicity and revealed favorable pharmacokinetics for a reasonable dosing schedule. These results pave the way for clinical trials with anti-cPD-1 in canine cancer patients to investigate predictive biomarkers and combination regimens to inform human clinical trials and bring a promising checkpoint inhibitor into the veterinary armamentarium.
Collapse
Affiliation(s)
- Sho Yoshimoto
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Ailian Xiong
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hong Wang
- Vetigenics LLC, B-Labs, Cira Center, Philadelphia, PA, USA
| | | | - Gayathri Gulendran
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Glassman
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, Philadelphia, PA, USA
| | - Don L. Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicola J. Mason
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
43
|
Jin X, Ma X, Zhao D, Yang L, Ma N. Immune microenvironment and therapeutic progress of recurrent hepatocellular carcinoma after liver transplantation. Transl Oncol 2022; 28:101603. [PMID: 36542991 PMCID: PMC9794975 DOI: 10.1016/j.tranon.2022.101603] [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: 10/24/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
HCC is a highly lethal tumor, and orthotopic liver transplantation, as one of the radical treatment methods for HCC, has opened-up a new therapeutic approach for the treatment of primary liver cancer. However, tumor recurrence after liver transplantation is the main reason that affects the long-term survival of recipients. At present, the application of ICIs has brought dawn to patients with refractory HCC. However, because of the special immune tolerance state created by long-term oral immunosuppressants in patients with HCC after liver transplantation, the current focus is how to regulate the immune balance of such patients and simultaneously maximize the anti-tumor effect. This article reviews the relationship between liver cancer and immunity, immune tolerance of liver transplantation, immune microenvironment after liver transplantation for HCC, and the application of immunotherapy in the recurrence of liver transplantation for HCC.
Collapse
Affiliation(s)
- Xin Jin
- Division of Liver Surgery and Organ Transplantation Center, Shenzhen Third People's Hospital, Second Affiliated Hospital of Southern University of Science and Technology, National Clinical Research Center for Infectious Disease, No.29 Bulan Road, Longgang District, Shenzhen, 518112, Guangdong Province, China
| | - Xiaoting Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Dong Zhao
- Division of Liver Surgery and Organ Transplantation Center, Shenzhen Third People's Hospital, Second Affiliated Hospital of Southern University of Science and Technology, National Clinical Research Center for Infectious Disease, No.29 Bulan Road, Longgang District, Shenzhen, 518112, Guangdong Province, China
| | - Lin Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China,Corresponding authors.
| | - Nan Ma
- Division of Liver Surgery and Organ Transplantation Center, Shenzhen Third People's Hospital, Second Affiliated Hospital of Southern University of Science and Technology, National Clinical Research Center for Infectious Disease, No.29 Bulan Road, Longgang District, Shenzhen, 518112, Guangdong Province, China,Corresponding authors.
| |
Collapse
|
44
|
Liu G, Lu J, Lim HS, Jin JY, Lu D. Applying interpretable machine learning workflow to evaluate exposure-response relationships for large-molecule oncology drugs. CPT Pharmacometrics Syst Pharmacol 2022; 11:1614-1627. [PMID: 36193885 PMCID: PMC9755920 DOI: 10.1002/psp4.12871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 11/05/2022] Open
Abstract
The application of logistic regression (LR) and Cox Proportional Hazard (CoxPH) models are well-established for evaluating exposure-response (E-R) relationship in large molecule oncology drugs. However, applying machine learning (ML) models on evaluating E-R relationships has not been widely explored. We developed a workflow to train regularized LR/CoxPH and tree-based XGboost (XGB) models, and derive the odds ratios for best overall response and hazard ratios for overall survival, across exposure quantiles to evaluate the E-R relationship using clinical trial datasets. The E-R conclusions between LR/CoxPH and XGB models are overall consistent, and largely aligned with historical pharmacometric analyses findings. Overall, applying this interpretable ML workflow provides a promising alternative method to assess E-R relationships for impacting key dosing decisions in drug development.
Collapse
Affiliation(s)
- Gengbo Liu
- Department of Clinical PharmacologyGenentechSouth San FranciscoCaliforniaUSA
| | - James Lu
- Department of Clinical PharmacologyGenentechSouth San FranciscoCaliforniaUSA
| | - Hong Seo Lim
- Department of Clinical PharmacologyGenentechSouth San FranciscoCaliforniaUSA
| | - Jin Yan Jin
- Department of Clinical PharmacologyGenentechSouth San FranciscoCaliforniaUSA
| | - Dan Lu
- Department of Clinical PharmacologyGenentechSouth San FranciscoCaliforniaUSA
| |
Collapse
|
45
|
Mallardo D, Giannarelli D, Vitale MG, Galati D, Trillò G, Esposito A, Isgrò MA, D'Angelo G, Festino L, Vanella V, Trojaniello C, White A, De Cristofaro T, Bailey M, Pignata S, Caracò C, Petrillo A, Muto P, Maiolino P, Budillon A, Warren S, Cavalcanti E, Ascierto PA. Nivolumab serum concentration in metastatic melanoma patients could be related to outcome and enhanced immune activity: a gene profiling retrospective analysis. J Immunother Cancer 2022; 10:jitc-2022-005132. [PMID: 36424033 PMCID: PMC9693654 DOI: 10.1136/jitc-2022-005132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Nivolumab is an anti-PD-1 antibody approved for treating metastatic melanoma (MM), for which still limited evidence is available on the correlation between drug exposure and patient outcomes. METHODS In this observational retrospective study, we assessed whether nivolumab concentration is associated with treatment response in 88 patients with MM and if the patient's genetic profile plays a role in this association. RESULTS We observed a statistically significant correlation between nivolumab serum concentration and clinical outcomes, measured as overall and progression-free survival. Moreover, patients who achieved a clinical or partial response tended to have higher levels of nivolumab than those who reached stable disease or had disease progression. However, the difference was not statistically significant. In particular, patients who reached a clinical response had a significantly higher concentration of nivolumab and presented a distinct genetic signature, with more marked activation of ICOS and other genes involved in effector T-cells mediated proinflammatory pathways. CONCLUSIONS In conclusion, these preliminary results show that in patients with MM, nivolumab concentration correlates with clinical outcomes and is associated with an increased expression of ICOS and other genes involved in the activation of T effectors cells.
Collapse
Affiliation(s)
| | | | | | - Domenico Galati
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Giusy Trillò
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Assunta Esposito
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Grazia D'Angelo
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Lucia Festino
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Vito Vanella
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Andrew White
- NanoString Technologies Inc, Seattle, Washington, USA
| | | | | | - Sandro Pignata
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Corrado Caracò
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | - Paolo Muto
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Piera Maiolino
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Alfredo Budillon
- Instituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Sarah Warren
- NanoString Technologies Inc, Seattle, Washington, USA
| | | | | |
Collapse
|
46
|
Poon V, Lu D. Performance of Cox proportional hazard models on recovering the ground truth of confounded exposure-response relationships for large-molecule oncology drugs. CPT Pharmacometrics Syst Pharmacol 2022; 11:1511-1526. [PMID: 35988264 PMCID: PMC9662202 DOI: 10.1002/psp4.12859] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 12/20/2022] Open
Abstract
A Cox proportional hazard (CoxPH) model is conventionally used to assess exposure-response (E-R), but its performance to uncover the ground truth when only one dose level of data is available has not been systematically evaluated. We established a simulation workflow to generate realistic E-R datasets to assess the performance of the CoxPH model in recovering the E-R ground truth in various scenarios, considering two potential reasons for the confounded E-R relationship. We found that at high doses, when the pharmacological effects are largely saturated, missing important confounders is the major reason for inferring false-positive E-R relationships. At low doses, when a positive E-R slope is the ground truth, either missing important confounders or mis-specifying the interactions can lead to inaccurate estimates of the E-R slope. This work constructed a simulation workflow generally applicable to clinical datasets to generate clinically relevant simulations and provide an in-depth interpretation on the E-R relationships with confounders inferred by the conventional CoxPH model.
Collapse
Affiliation(s)
- Victor Poon
- Modeling and Simulation Group, Department of Clinical PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dan Lu
- Modeling and Simulation Group, Department of Clinical PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
47
|
Koguchi Y, Redmond WL. A Novel Class of On-Treatment Cancer Immunotherapy Biomarker: Trough Levels of Antibody Therapeutics in Peripheral Blood. Immunol Invest 2022; 51:2159-2175. [PMID: 36301695 DOI: 10.1080/08820139.2022.2131570] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
While immune checkpoint blockade has revolutionized cancer treatment, unfortunately most patients do not benefit from this treatment. Many pharmacodynamic (PD) studies have revealed essential requirements for successful cancer immunotherapy that may provide insight into how we can improve these agents. Despite enormous efforts focused on interrogating the immune system using different biospecimens (e.g. blood, primary tumor, metastatic tumor, microbiome samples), a variety of technologies (e.g. flow cytometry, bulk and single-cell RNA-sequencing, immunohistochemistry), and wide-ranging disciplines (e.g. pathology, genomics, bioinformatics, immunology, cancer biology, metabolomics, bacteriology), discovery of consistent biomarkers of response have remained elusive. Pharmacokinetics (PK) studies, however, not only provide critical information regarding safe dosing but may also reveal useful biomarkers. For example, recent studies found that trough levels of therapeutic monoclonal antibodies (mAbs) or clearance (CL) of them were associated with clinical outcome, which suggests that trough levels of mAbs may represent a new class of on-treatment cancer immunotherapy biomarker. In this review, we summarize the potential utility of trough levels of mAbs, the mechanism of varying PK, consideration for therapeutic drug monitoring, and assay attributes that will facilitate wider utilization of PK information in conjunction with PD assessments.
Collapse
Affiliation(s)
- Yoshinobu Koguchi
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| |
Collapse
|
48
|
Optimizing the dose and schedule of immune checkpoint inhibitors in cancer to allow global access. Nat Med 2022; 28:2236-2237. [PMID: 36202999 DOI: 10.1038/s41591-022-02029-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
49
|
Jin X, Zhang K, Fang T, Zeng X, Yan X, Tang J, Liang Z, Xie L, Zhao D. Low-dose PD-1 inhibitor combined with lenvatinib for preemptive treatment of recurrence after liver transplantation for hepatocellular carcinoma: Case report and literature review. Front Oncol 2022; 12:951303. [PMID: 36119543 PMCID: PMC9478730 DOI: 10.3389/fonc.2022.951303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/10/2022] [Indexed: 11/18/2022] Open
Abstract
Orthotopic liver transplantation (OLT), as one of the curative methods for the treatment of hepatocellular carcinoma (HCC), has brought hope to patients with HCC. However, treatment options for HCC recurrence and metastasis after liver transplantation are limited. Immune checkpoint inhibitor (ICI), such as programmed cell death protein 1 (PD-1) inhibitor, have been successfully used in advanced or metastatic HCC, but the data on the safety of PD-1 inhibitor after liver transplantation is limited. In this article, we report a 47-year-old patient with acute-on-chronic liver failure and multiple HCC who was successfully treated with liver transplantation. On the 45th day after OLT, the patient’s alpha fetoprotein (AFP) and lens culinaris agglutinin-reactive fraction of AFP (AFP-L3) were increased, and imaging examination showed no residual tumor. The patient had high risk factors for tumor recurrence before operation, so the possibility of tumor recurrence was considered. When the tumor markers showed an upward trend, we immediately treated the patient with lenvatinib 8 mg, after half a month, the AFP and AFP-L3 continued to increase compared with before. Then we used low-dose nivolumab 40mg, the patient’s AFP and AFP-L3 gradually decreased. One month later, a second low-dose nivolumab 40mg was given, and the patient’s tumor markers gradually decreased to normal. No acute rejection and other complications occurred during the treatment. So far, we have followed up this patient for 2 years, and no tumor recurrence was observed. To our knowledge, this is the first reported case using a low dose of nivolumab in combination with lenvatinib to prevent recurrence of HCC after liver transplantation.
Collapse
|
50
|
Cheng Y, Hong K, Chen N, Yu X, Peluso T, Zhou S, Li Y. Aiding early clinical drug development by elucidation of the relationship between tumor growth inhibition and survival in relapsed/refractory multiple myeloma patients. EJHAEM 2022; 3:815-827. [PMID: 36051011 PMCID: PMC9422038 DOI: 10.1002/jha2.494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
Early prognosis of clinical efficacy is an urgent need for oncology drug development. Herein, we systemically examined the quantitative approach of tumor growth inhibition (TGI) and survival modeling in the space of relapsed and refractory multiple myeloma (MM), aiming to provide insights into clinical drug development. Longitudinal serum M-protein and progression-free survival (PFS) data from three phase III studies (N = 1367) across six treatment regimens and different patient populations were leveraged. The TGI model successfully described the longitudinal M-protein data in patients with MM. The tumor inhibition and growth parameters were found to vary as per each study, likely due to the patient population and treatment regimen difference. Based on a parametric time-to-event model for PFS, M-protein reduction at week 4 was identified as a significant prognostic factor for PFS across the three studies. Other factors, including Eastern Cooperative Oncology Group performance status, prior anti-myeloma therapeutics, and baseline serum ß2-microglobulin level, were correlated with PFS as well. In conclusion, patient disease characteristics (i.e., baseline tumor burden and treatment lines) were important determinants of tumor inhibition and PFS in MM patients. M-protein change at week 4 was an early prognostic biomarker for PFS.
Collapse
Affiliation(s)
- Yiming Cheng
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| | - Kevin Hong
- Global Drug DevelopmentBristol Myers SquibbNew JerseyUSA
| | - Nianhang Chen
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| | - Xin Yu
- Global Biometric SciencesBristol Myers SquibbNew JerseyUSA
| | - Teresa Peluso
- Global Drug Development Bristol Myers SquibbBoudrySwitzerland
| | - Simon Zhou
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| | - Yan Li
- Clinical Pharmacology & PharmacometricsBristol Myers SquibbNew JerseyUSA
| |
Collapse
|