1
|
DeBonis J, Igoshin OA. How does mathematical modeling improve our understanding and design of biological immunomodulators? Ther Deliv 2025:1-4. [PMID: 40383964 DOI: 10.1080/20415990.2025.2506980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025] Open
Affiliation(s)
| | - Oleg A Igoshin
- Department of Bioengineering, Rice University, Houston, TX, USA
- Center for Theoretical Biological Physics, Department of Chemistry, Department of Biosciences, Rice University, Houston, TX, USA
| |
Collapse
|
2
|
Jeon WJ, Son SY, Abodunrin F, Khanna A, Patel JD, Thawani R. Dosing immune-checkpoint inhibitors: Opportunities for the future. Curr Probl Cancer 2025; 57:101204. [PMID: 40339394 DOI: 10.1016/j.currproblcancer.2025.101204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
With recent breakthroughs in immunotherapy, particularly with the approval of immune checkpoint inhibitors for various cancer indications, patients now have a wider array of treatment options, even for those with metastatic disease. Still, the survival benefit of immune-checkpoint inhibitors is modest, and there is concern about drug toxicity. In addition, there is ongoing exploration into combination therapy involving immune-checkpoint inhibitors, which come at the risk of increased toxicity. Unfortunately, due to the cost of the currently approved doses and dosing intervals, many patients in the community in the United States and low- and middle-income countries lack access to these transformative therapies. Further, the observation of resistance to immune-checkpoint inhibitors and limitations of currently approved doses and dosing intervals warrants changes in current practice. This review paper discusses both model-based and clinical studies in the current literature. Strategies for improving access to immune-checkpoint inhibitors and expanding their utilization, including weight-based dosing instead of fixed dosing, dose and dose interval adjustments, development of biomarkers and scoring systems for personalization of immune-checkpoint inhibitors, and alternative trial design, are discussed.
Collapse
Affiliation(s)
- Won Jin Jeon
- Division of Hematology & Oncology, Loma Linda University, Loma Linda, CA, USA
| | - So Young Son
- Loma Linda University School of Pharmacy, Loma Linda University, Loma Linda, CA, USA
| | - Faith Abodunrin
- Biological Sciences Division, University of Chicago, Chicago, IL, USA
| | | | | | - Rajat Thawani
- Division of Hematology & Oncology, Knight Cancer Institute Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
3
|
Kondo S, Kitano S, Sato J, Katsuya Y, Kogawa T, Mizugaki H, Miyamoto I, Iwasawa S, Imaizumi T, Tomita H, Murakami C, Miyake T, Yamamoto N. An investigation of extended-interval dosing of atezolizumab in Japanese patients with advanced solid tumors: safety and pharmacokinetics of a dose of 1680 mg every 4 weeks. Invest New Drugs 2025:10.1007/s10637-024-01498-0. [PMID: 40310568 DOI: 10.1007/s10637-024-01498-0] [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: 12/11/2024] [Accepted: 12/30/2024] [Indexed: 05/02/2025]
Abstract
In Japan, atezolizumab is indicated for several cancers at a dose of 1200 mg every 3 weeks or 840 mg every 2 weeks. This open-label study (jRCT2031220151) aimed to assess an atezolizumab monotherapy dose of 1680 mg every 4 weeks (Q4W) in Japanese patients ≥ 18 years of age with advanced or recurrent solid tumors that were not responsive to standard treatment. The primary endpoints were tolerability, safety, and pharmacokinetics (PK). Secondary endpoints included overall response rate and progression-free survival. Overall, 21 patients were enrolled in the study. The median age for males (42.9%) and females (57.1%) was 61 years, and the median (range) treatment duration was 29.0 (1-224) days. During the dose-limiting toxicity (DLT) evaluation period, 3 out of 6 (50.0%) patients experienced at least 1 adverse event, although no DLTs or deaths were experienced. The PK profile of atezolizumab 1680 mg Q4W monotherapy in cycle 1 after 30 min of administration had an arithmetic mean maximum concentration (standard deviation [SD]) of 699 (146) µl/mL and a mean minimum concentration (SD) 133 (46.0) µl/mL, The mean (SD) area under the curve was 7180 (1340) days‧µg/mL. These data show that atezolizumab 1680 mg Q4W monotherapy was well tolerated in Japanese patients with no new safety concerns, suggesting that this less frequent dosing regimen could have the potential to offer greater flexibility and convenience for patients and caregivers.
Collapse
Affiliation(s)
- Shunsuke Kondo
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan.
- Department of Medical Oncology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Shigehisa Kitano
- Department of Advanced Medical Development, The Cancer Institute Hospital of JFCR, Tokyo, Japan.
| | - Jun Sato
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Yuki Katsuya
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| | - Takahiro Kogawa
- Department of Advanced Medical Development, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Hidenori Mizugaki
- Department of Advanced Medical Development, The Cancer Institute Hospital of JFCR, Tokyo, Japan
- Department of Respiratory Medicine, NHO Hokkaido Cancer Center, Sapporo, Japan
| | - Ippei Miyamoto
- Department of Advanced Medical Development, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Shunichiro Iwasawa
- Oncology Lifecycle Management Department, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Tatsuki Imaizumi
- Biometrics Department, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Hiroaki Tomita
- Biometrics Department, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Chika Murakami
- Oncology Clinical Development Department, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Takeshi Miyake
- Pharmaceutical Science Department, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Noboru Yamamoto
- Department of Experimental Therapeutics, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
4
|
Rizwan A, Giovinazzo H, Yu T, Gao Y, Wang K, Xu F, Wan Y, Wang J, Sahasranaman S, Campbell M, Schnell P, Abdrashitov R, Hanley WD, Budha N. Alternative Dosing Regimens of Tislelizumab Using a Pharmacometrics Model-Based Approach. Clin Transl Sci 2025; 18:e70223. [PMID: 40286320 PMCID: PMC12033006 DOI: 10.1111/cts.70223] [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: 01/07/2025] [Revised: 03/18/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025] Open
Abstract
Tislelizumab 200 mg once every 3 weeks (Q3W) is approved for the treatment of multiple cancers. We used a model-based approach to propose three alternative dosing regimens, 150 mg Q2W, 300 mg Q4W, and 400 mg Q6W, with the aims of providing flexible treatment regimens compatible with background chemotherapy and/or reducing infusion visits. A previously developed population pharmacokinetic model was used to simulate pharmacokinetic exposure of the alternative regimens. Regulatory guidance on alternative dosing was used to define pharmacokinetics-based criteria. Alternative doses were selected by simulation, exposure matching, and comparison to the reference regimen of 200 mg Q3W. Deviations from pharmacokinetics-based criteria were bridged using appropriate safety and efficacy references and exposure-response analyses. All three alternative dosing regimens produced comparable exposures versus 200 mg Q3W. Although simulated peak serum concentration (Cmax) at 300 mg Q4W and 400 mg Q6W was higher versus 200 mg Q3W, this was below the Cmax of the 5 mg/kg Q3W safety reference. And while the trough serum concentration (Ctrough) for 400 mg Q6W was slightly lower versus 200 mg Q3W, it was 10.7% higher than the 2 mg/kg efficacy reference Ctrough, and therefore, within the concentration range in which a flat exposure-efficacy relationship of tislelizumab has been established. Tislelizumab regimens of 150 mg Q2W, 300 mg Q4W, and 400 mg Q6W are expected to result in similar safety and efficacy as 200 mg Q3W.
Collapse
Affiliation(s)
- Ahsan Rizwan
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Hugh Giovinazzo
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Tian Yu
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Yuying Gao
- PharmacometricsShanghai Qiangshi Information Technology Co., Ltd.ShanghaiChina
| | - Kun Wang
- PharmacometricsShanghai Qiangshi Information Technology Co., Ltd.ShanghaiChina
| | - Fengyan Xu
- PharmacometricsShanghai Qiangshi Information Technology Co., Ltd.ShanghaiChina
| | - Ya Wan
- Scientific ProgrammingBeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Jun Wang
- Global Statistics and Data SciencesBeiGene (Beijing), Co., Ltd.BeijingChina
| | | | | | | | | | - William D. Hanley
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Nageshwar Budha
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| |
Collapse
|
5
|
Jogiraju V, Pawar P, Yager J, Ling J, Shen G, Chiu A, Hughes E, Palaparthy R, Carter C, Singh R. Pharmacokinetics and safety of once-yearly lenacapavir: a phase 1, open-label study. Lancet 2025; 405:1147-1154. [PMID: 40086460 DOI: 10.1016/s0140-6736(25)00405-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Long-acting antiretrovirals can address barriers to HIV pre-exposure prophylaxis (PrEP), such as stigma and adherence. In two phase 3 trials, twice-yearly subcutaneous lenacapavir was safe and highly efficacious for PrEP in diverse populations. Furthering long-acting PrEP efforts, this study assessed the pharmacokinetics and safety of two once-yearly intramuscular lenacapavir formulations. METHODS This phase 1, open-label study in participants aged 18-55 years without HIV evaluated the pharmacokinetics, safety, and tolerability of two lenacapavir free acid formulations administered by ventrogluteal intramuscular injection as a single 5000 mg dose (formulation 1 with 5% w/w ethanol, formulation 2 with 10% w/w ethanol). Pharmacokinetic samples were collected at prespecified timepoints up to 56 weeks. Lenacapavir plasma concentrations were measured with a validated liquid chromatography-tandem mass spectrometry method and summarised with non-compartmental analysis. Pharmacokinetic parameters evaluated included the area under the concentration-time curve for the once-yearly dosing interval calculated from days 1 to 365 (AUCdays 1-365), peak plasma concentration, time to reach peak plasma concentration, and trough concentration (Ctrough). Plasma concentration data from phase 3 studies of twice-yearly subcutaneous lenacapavir (PURPOSE 1 and PURPOSE 2) were pooled for comparison with once-yearly intramuscular lenacapavir formulations. Safety and tolerability, including participant-reported pain scores, were assessed. FINDINGS 20 participants received lenacapavir formulation 1 and 20 received lenacapavir formulation 2. For estimation of pharmacokinetic parameters, sample size varied over time with at least 13 participants (formulation 1) and at least 19 participants (formulation 2) due to early discontinuations for reasons unrelated to the study drug. Following administration of intramuscular lenacapavir, concentrations increased rapidly, and median time to maximum concentration was 84·1 days (IQR 56·1-112·0) for formulation 1 and 69·9 days (55·3-105·5) for formulation 2. The highest median concentration of once-yearly intramuscular lenacapavir (247·0 ng/mL [IQR 184·0-346·0] for formulation 1, 336·0 ng/mL [233·5-474·3] for formulation 2) remained above the highest median twice-yearly subcutaneous lenacapavir concentration (67·3 ng/mL [46·8-91·4]). Median Ctrough at the end of 52 weeks for formulation 1 was 57·0 ng/mL (IQR 49·9-72·4) and for formulation 2 was 65·6 ng/mL (41·8-87·1), exceeding the median twice-yearly subcutaneous lenacapavir Ctrough of 23·4 ng/mL (15·7-34·3) at the end of 26 weeks. Median AUCdays 1-365 for formulation 1 was 1011·1 h*μg/mL (IQR 881·0-1490·2) and for formulation 2 was 1274·0 h*μg/mL (1177·3-1704·8). Adverse events were mostly grade 1 or 2. The most common was injection-site pain (16 [80%] participants given formulation 1, 15 [75%] given formulation 2), which was generally mild, resolved within 1 week, and was substantially reduced by pretreatment with ice. INTERPRETATION Following administration of once-yearly intramuscular lenacapavir, median plasma concentrations exceeded those associated with efficacy in phase 3 studies of twice-yearly subcutaneous lenacapavir for PrEP for at least 56 weeks. Both formulations were safe and well tolerated. These data show the potential for biomedical HIV prevention with a once-yearly dosing interval. FUNDING Gilead Sciences.
Collapse
Affiliation(s)
| | | | | | - John Ling
- Gilead Sciences, Foster City, CA, USA
| | - Gong Shen
- Gilead Sciences, Foster City, CA, USA
| | - Anna Chiu
- Gilead Sciences, Foster City, CA, USA
| | | | | | | | | |
Collapse
|
6
|
Lee N, Rhee S, Koo SM, Kim SW, Lee GE, Yie YA, Kim Y. Navigating Recent Changes in Dosing Information: Dynamics of FDA-Approved Monoclonal Antibodies in Post-Marketing Realities. Clin Transl Sci 2025; 18:e70125. [PMID: 39807701 PMCID: PMC11729449 DOI: 10.1111/cts.70125] [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: 07/15/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025] Open
Abstract
Monoclonal antibodies (mAbs) are critical components in the therapeutic landscape, but their dosing strategies often evolve post-approval as new data emerge. This review evaluates post-marketing label changes in dosing information for FDA-approved mAbs from January 2015 to September 2024, with a focus on both initial and extended indications. We systematically analyzed dosing modifications, categorizing them into six predefined groups: Dose increases or decreases, inclusion of new patient populations by body weight or age, shifts from body weight-based dosing to fixed regimens, and adjustments in infusion rates. Among the 86 mAbs evaluated, 21% (n = 18) exhibited changes in dosing information for the initial indication, with a median time to modification of 37.5 months (range: 5-76 months). Furthermore, for mAbs with extended indications (n = 26), 19.2% (n = 5) underwent dosing changes in their first extensions, with a median time to adjustment of 31 months (range: 8-71 months). Key drivers for these adjustments included optimizing therapeutic efficacy, addressing safety concerns, accommodating special populations, and enhancing patient convenience. We also discuss the role of model-informed drug development, real-world evidence, and pharmacogenomics in refining mAb dosing strategies. These insights underscore the importance of ongoing monitoring and data integration in the post-marketing phase, providing a foundation for future precision medicine approaches in mAb therapy.
Collapse
Affiliation(s)
- Nai Lee
- College of Pharmacy, Daegu Catholic UniversityGyeongsanKorea
| | - Su‐jin Rhee
- College of Pharmacy, Wonkwang UniversityIksanKorea
| | - Seong Min Koo
- College of Pharmacy, Daegu Catholic UniversityGyeongsanKorea
| | - So Won Kim
- College of Pharmacy, Daegu Catholic UniversityGyeongsanKorea
| | - Gyo Eun Lee
- College of Pharmacy, Daegu Catholic UniversityGyeongsanKorea
| | - Yoon A Yie
- College of Pharmacy, Daegu Catholic UniversityGyeongsanKorea
| | - Yun Kim
- College of Pharmacy, Daegu Catholic UniversityGyeongsanKorea
| |
Collapse
|
7
|
Barrett PM, Daly FP, Zhang M, O'Reilly A, Heckmann P, Tierney P, Murray D, Mullooly M, Bennett K. The impact of the COVID-19 pandemic on the dispensing of systemic anti-cancer therapy (SACT) in Ireland: A population based study. PUBLIC HEALTH IN PRACTICE 2024; 8:100557. [PMID: 39635543 PMCID: PMC11615519 DOI: 10.1016/j.puhip.2024.100557] [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: 03/06/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
Objectives The COVID-19 pandemic had considerable implications for cancer related care. This study aimed to examine its impact on the dispensing of systemic anti-cancer therapy (SACT) in Ireland. Study design A repeated cross-sectional design was used which involved a quasi-experimental interrupted time series analysis (ITSA), and autoregressive integrated moving average (ARIMA) models. Methods This nationally representative study utilised monthly pharmacy claims (i.e. dispensing) data from community and hospital schemes. Dispensed items among individuals prescribed any SACT from January 2019 to April 2021 were included. Results During the study period, 641,273 SACT items were dispensed, including 57,199 chemotherapeutic agents (8.9 %), 15,970 immunotherapeutic agents (2.5 %), 87,813 targeted therapies (13.7 %), and 480,291 (74.9 %) endocrine therapies. There were on average 3.3 and 4.1 fewer immunotherapy and targeted therapy agents, respectively, dispensed per 100,000 population per month post-March 2020 (vs. expected), compared to the level prior to March 2020. For endocrine therapy, there was a significant slowing of the trend post-March 2020 compared to the pre-pandemic period (slope change = -1.72, 95 % CI -2.9 to -0.5; p<0.01). Conclusion There was a significant level decrease in the dispensing of immunotherapy and targeted therapy during the first year of the pandemic, and a slowing of the trend for endocrine therapies. However, no differences in the dispensing of other SACT were observed.
Collapse
Affiliation(s)
- Peter M. Barrett
- Department of Public Health HSE South-West, St. Finbarr’s Hospital, Cork, Ireland
- School of Public Health, University College Cork, Cork, Ireland
| | - Fionn P. Daly
- School of Public Health, University College Cork, Cork, Ireland
- School of Medicine and Health, University College Cork, Cork, Ireland
| | - Mengyang Zhang
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Patricia Heckmann
- HSE National Cancer Control Programme, Kings Inn House, Dublin, Ireland
| | | | - Deirdre Murray
- School of Public Health, University College Cork, Cork, Ireland
- National Cancer Registry in Ireland, Cork, Ireland
| | - Maeve Mullooly
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Kathleen Bennett
- School of Population Health, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| |
Collapse
|
8
|
Cohen G, Rapoport B, Chan SW, Ruff P, Arance A, Mujika Eizmendi K, Houghton B, Brown MP, Zielinski RM, Muñoz Couselo E, Lyle M, Anderson JR, Jain L, de Alwis D, Lala M, Akala O, Chartash E, Jacobs C. Pembrolizumab 400 mg every 6 weeks as first-line therapy for advanced melanoma (KEYNOTE-555): Results from cohort B of an open-label, phase 1 study. PLoS One 2024; 19:e0309778. [PMID: 39531423 PMCID: PMC11556718 DOI: 10.1371/journal.pone.0309778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/02/2024] [Indexed: 11/16/2024] Open
Abstract
Intravenous pembrolizumab 400 mg every 6 weeks was approved across tumor types based on pharmacokinetic modeling, which showed exposures consistent with previous standard dosing of 200 mg or 2 mg/kg every 3 weeks, and early results of cohort B of the phase 1 KEYNOTE-555 study. Results after ≥1 year of potential follow-up for all patients in cohort B of KEYNOTE-555 are presented. Patients aged ≥18 years with previously untreated stage III/IV melanoma received pembrolizumab 400 mg every 6 weeks for ≤18 cycles. The primary endpoint was objective response rate per RECIST v1.1 by blinded independent central review. Secondary endpoints included duration of response, progression-free survival, pharmacokinetics, and safety. Overall, 101 patients received pembrolizumab. Median projected follow-up was 21.9 months (range, 17.0-25.7). The objective response rate was 50.5% (95% CI: 40.4-60.6; 19 complete responses, 32 partial responses). Median duration of response was not reached (NR; range, 2.4+ to 21.0+ months). Median progression-free survival was 13.8 months (95% CI: 4.1-NR). Observed pharmacokinetic exposures were consistent with model predictions for pembrolizumab 400 mg every 6 weeks and other approved and tested schedules (2 mg/kg or 200 mg every 3 weeks). Grade 3-4 treatment-related adverse events occurred in 13 patients (12.9%). No deaths were considered treatment related. These results support the pharmacokinetic modeling and demonstrate that the benefit-risk profile of pembrolizumab 400 mg Q6W is consistent with that of 200 mg or 2 mg/kg every 3 weeks. Clinically meaningful objective response rate and durable progression-free survival within the expected range for first-line pembrolizumab were observed. Clinical trial registry: ClinicalTrials.gov, NCT03665597.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Melanoma/drug therapy
- Melanoma/pathology
- Melanoma/mortality
- Middle Aged
- Female
- Male
- Aged
- Adult
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Aged, 80 and over
- Progression-Free Survival
- Cohort Studies
- Drug Administration Schedule
- Treatment Outcome
Collapse
Affiliation(s)
- Graham Cohen
- Mary Potter Oncology Centre, Pretoria, South Africa
| | - Bernardo Rapoport
- The Medical Oncology Centre of Rosebank, Johannesburg, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Sze W. Chan
- Sandton Oncology, Johannesburg, South Africa
| | - Paul Ruff
- University of Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa
| | - Ana Arance
- Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Baerin Houghton
- Port Macquarie Base Hospital, Port Macquarie, New South Wales, Australia
| | - Michael P. Brown
- Cancer Clinical Trials Unit, Royal Adelaide Hospital, and School of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Robert M. Zielinski
- Central West Cancer Care Centre, Orange Hospital, Orange, New South Wales, Australia
- Western Sydney University, Sydney, New South Wales, Australia
| | - Eva Muñoz Couselo
- Department of Medical Oncology, Melanoma and Other Skin Cancers Unit, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Megan Lyle
- Liz Plummer Cancer Care Centre, Cairns, Queensland, Australia
| | | | - Lokesh Jain
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Dinesh de Alwis
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Mallika Lala
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Omobolaji Akala
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Elliot Chartash
- Merck & Co., Inc., Rahway, New Jersey, United States of America
| | - Conrad Jacobs
- Cancer Care: Clinical & Radiation Oncology, Cape Town, South Africa
| |
Collapse
|
9
|
Vora B, Jindal A, Velasquez E, Lu J, Wu B. Integrating real-world data and machine learning: A framework to assess covariate importance in real-world use of alternative intravenous dosing regimens for atezolizumab. Clin Transl Sci 2024; 17:e70077. [PMID: 39558509 PMCID: PMC11573720 DOI: 10.1111/cts.70077] [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: 06/12/2024] [Revised: 10/08/2024] [Accepted: 11/04/2024] [Indexed: 11/20/2024] Open
Abstract
The increase in the availability of real-world data (RWD), in combination with advances in machine learning (ML) methods, provides a unique opportunity for the integration of the two to explore complex clinical pharmacology questions. Here we present a recently developed RWD/ML framework that utilizes ML algorithms to understand the influence and importance of various covariates on the use of a given dose and schedule for drugs that have multiple approved dosing regimens. To demonstrate the application of this framework, we present atezolizumab as a use case on account of its three approved alternative intravenous (IV) dosing regimens. As expected, the real-world use of atezolizumab has generally been increasing since 2016 for the 1200 mg every 3 weeks regimen and since 2019 for the 1680 mg every 4 weeks regimen. Out of the ML algorithms evaluated, XGBoost performed the best, as measured by the area under the precision-recall curve, with an emphasis on the under-sampled class given the imbalance in the data. The importance of features was measured by Shapley Additive exPlanations (SHAP) values and showed metastatic breast cancer and use of protein-bound paclitaxel as the most correlated with the use of 840 mg every 2 weeks. Although patient usage data for alternative IV dosing regimens are still maturing, these analyses provide initial insights on the use of atezolizumab and set up a framework for the re-analysis of atezolizumab (at a future data cut) as well as application to other molecules with approved alternative dosing regimens.
Collapse
Affiliation(s)
- Bianca Vora
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Ashutosh Jindal
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Erick Velasquez
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - James Lu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
10
|
Ngo HX, Oh E, Li C, Yu J. Oncology Dose Selection in Subsequent Indications: What Can We Learn From FDA-approved Oncology Drugs? Clin Ther 2024; 46:927-937. [PMID: 39304367 DOI: 10.1016/j.clinthera.2024.08.020] [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: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE The modern oncology drug development landscape has shifted away from traditional cytotoxic chemotherapies. Following their initial approvals, many oncology drugs have been approved in subsequent indications either as monotherapy or in combination to benefit a broader patient population. To date, dose selection strategies for subsequent indications have not been systematically reviewed. This review examines how approved dosing regimens were selected in subsequent indications for FDA-approved oncology drugs. METHODS The Drugs@FDA database was used to identify FDA-approved new molecular entities (NMEs) between 2010 and 2023. NMEs with more than 1 approved indication were included in the analysis. In total, the dosing regimens for 67 novel oncology drugs that obtained FDA approvals for multiple indications were evaluated. FINDINGS Overall, in subsequent indications, 72% of NMEs used the same or clinically equivalent alternative dosing regimens to those approved in the initial indications. Amongst the 28% of NMEs that used different dosing regimens, safety/tolerability was the leading cause of a dosing regimen changes in both monotherapy and combination therapy settings. Other factors leading to changes in dosing regimens include differences in tumor biology, disease burden, pharmacokinetics, and overall benefit-risk profiles obtained from dose-finding studies. IMPLICATIONS Our analysis highlighted the importance of selecting a safe, tolerable, and yet efficacious dosing regimen for the initial indication as a suboptimal initially approved regimen could lead to dosing regimen changes in later indications. Preclinical and clinical data could be leveraged to understand the pharmacology, pharmacokinetic, and pharmacodynamic differences between indications and thus support dose selection in subsequent indications.
Collapse
Affiliation(s)
- Huy X Ngo
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Elise Oh
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Chunze Li
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA
| | - Jiajie Yu
- Department of Clinical Pharmacology, Genentech, Inc., South San Francisco, California, USA.
| |
Collapse
|
11
|
Milenković-Grišić AM, Hayes S, Farrell C, Kuroki Y, Bertolino M, Venkatakrishnan K, Girard P. Model-informed Evidence for Clinical Non-inferiority of Every-2-Weeks Versus Standard Weekly Dosing Schedule of Cetuximab in Metastatic Colorectal Cancer. Clin Pharmacol Ther 2024; 116:1071-1081. [PMID: 39132970 DOI: 10.1002/cpt.3345] [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: 11/24/2023] [Accepted: 05/30/2024] [Indexed: 08/13/2024]
Abstract
Cetuximab was initially developed and approved as a first-line treatment in patients with unresectable metastatic colorectal cancer (mCRC) for weekly administration (250 mg/m2 Q1W with 400 mg/m2 loading dose). An every-2-weeks schedule (500 mg/m2 Q2W) was approved recently by several health authorities. Being synchronized with chemotherapy, Q2W administration should improve patients' convenience and healthcare resource utilization. Herein, we present evidence of non-inferiority of Q2W cetuximab, compared with Q1W dosing using pharmacometrics modeling and clinical trial simulation (CTS). Pooled data from five phase I-III clinical trials in 852 patients with KRAS wild-type mCRC treated with Q1W or Q2W cetuximab were modeled using a population exposure-tumor size (TS) model linked to overall survival (OS); exposure was derived from a previously established population pharmacokinetic model. A semi-mechanistic TS model adapted from the Claret model incorporated killing rate proportional to cetuximab area under the concentration-time curve over 2 weeks (AUC) with Eastern Cooperative Oncology Group (ECOG) status as covariate on baseline TS. The OS was modeled with Weibull hazard using ECOG, baseline TS, primary tumor location, and predicted percent change in TS at 8 weeks as covariates. Model-based simulations revealed indistinguishable early tumor shrinkage and survival between Q2W vs. Q1W cetuximab. CTS evaluated OS non-inferiority (predefined margin of 1.25) in 1,000 trials, each with 2,000 virtual patients receiving Q2W or Q1W cetuximab (1:1), and demonstrated non-inferiority in 94% of cases. Taken together, these analyses provide model-based evidence for clinical non-inferiority of Q2W vs. Q1W cetuximab in mCRC with potential benefits to patients and healthcare providers.
Collapse
Affiliation(s)
| | | | | | - Yoshihiro Kuroki
- Merck Biopharma Co. Ltd. (an affiliate of Merck KGaA, Darmstadt, Germany), Tokyo, Japan
| | | | | | - Pascal Girard
- Merck Institute of Pharmacometrics, Ares Trading S.A. (an affiliate of Merck KGaA, Darmstadt, Germany), Lausanne, Switzerland
| |
Collapse
|
12
|
Soares Dos Santos MP, Bernardo RMC, Vidal J, Moreira A, Torres DFM, Herdeiro CAR, Santos HA, Gonçalves G. Next-generation chemotherapy treatments based on black hole algorithms: From cancer remission to chronic disease management. Comput Biol Med 2024; 180:108961. [PMID: 39106673 DOI: 10.1016/j.compbiomed.2024.108961] [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: 03/15/2024] [Revised: 07/10/2024] [Accepted: 07/26/2024] [Indexed: 08/09/2024]
Abstract
PROBLEM Therapeutic planning strategies have been developed to enhance the effectiveness of cancer drugs. Nevertheless, their performance is highly limited by the inefficient biological representativeness of predictive tumor growth models, which hinders their translation to clinical practice. OBJECTIVE This study proposes a disruptive approach to oncology based on nature-inspired control using realistic Black Hole physical laws, in which tumor masses are trapped to experience attraction dynamics on their path to complete remission or to become a chronic disease. This control method is designed to operate independently of individual patient idiosyncrasies, including high tumor heterogeneities and highly uncertain tumor dynamics, making it a promising avenue for advancing beyond the limitations of the traditional survival probabilistic paradigm. DESIGN Here, we provide a multifaceted study of chemotherapy therapeutic planning that includes: (1) the design of a pioneering controller algorithm based on physical laws found in the Black Holes; (2) investigation of the ability of this controller algorithm to ensure stable equilibrium treatments; and (3) simulation tests concerning tumor volume dynamics using drugs with significantly different pharmacokinetics (Cyclophosphamide and Atezolizumab), tumor volumes (200 mm3 and 12 732 mm3) and modeling characterizations (Gompertzian and Logistic tumor growth models). RESULTS Our results highlight the ability of this new astrophysical-inspired control algorithm to perform effective chemotherapy treatments for multiple tumor-treatment scenarios, including tumor resistance to chemotherapy, clinical scenarios modelled by time-dependent parameters, and highly uncertain tumor dynamics. CONCLUSIONS Our findings provide strong evidence that cancer therapy inspired by phenomena found in black holes can emerge as a disruptive paradigm. This opens new high-impacting research directions, exploring synergies between astrophysical-inspired control algorithms and Artificial Intelligence applied to advanced personalized cancer therapeutics.
Collapse
Affiliation(s)
- Marco P Soares Dos Santos
- Center for Mechanical Technology & Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal; Intelligent Systems Associate Laboratory (LASI), Guimarães, Portugal.
| | - Rodrigo M C Bernardo
- Center for Mechanical Technology & Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal
| | - JoãoV Vidal
- Department of Physics and Aveiro Institute of Materials (CICECO), University of Aveiro, Aveiro, Portugal; Department of Physics and Institute for Nanostructures, Nanomodelling and Nanofabrication (I3N), University of Aveiro, Aveiro, Portugal
| | - Ana Moreira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193, Aveiro, Portugal
| | - Delfim F M Torres
- Center for Research and Development in Mathematics and Applications (CIDMA), Department of Mathematics, University of Aveiro, Aveiro, Portugal
| | - Carlos A R Herdeiro
- Center for Research and Development in Mathematics and Applications (CIDMA), Department of Mathematics, University of Aveiro, Aveiro, Portugal
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Gil Gonçalves
- Center for Mechanical Technology & Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, Aveiro, Portugal; Intelligent Systems Associate Laboratory (LASI), Guimarães, Portugal
| |
Collapse
|
13
|
Yagishita S, Goto Y, Nishio M, Akamatsu H, Hayashi H, Miura S, Tamada K, Kagamu H, Hamada A, Ohuchi M, Gemma A, Yoshino I, Misumi T, Hata A, Hara S, Kijima T, Masaki F, Iwasawa S, Nakagawa S, Tatsuno M, Mitsudomi T. Real-World Pharmacokinetics, Effectiveness, and Safety of Atezolizumab in Patients With Unresectable Advanced or Recurrent NSCLC: An Exploratory Study of J-TAIL. JTO Clin Res Rep 2024; 5:100683. [PMID: 39091595 PMCID: PMC11293501 DOI: 10.1016/j.jtocrr.2024.100683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/15/2024] [Accepted: 05/26/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction This study validated real-world pharmacokinetic (PK) data using an established population PK (PopPK) model for atezolizumab in Japanese patients with NSCLC and explored the relationship between PK parameters, effectiveness, and adverse events (AEs) for the 1200 mg once every three weeks regimen. Methods A subgroup of 262 of 1039 patients from J-TAIL consented to this exploratory research for PK evaluation of atezolizumab monotherapy for unresectable advanced/recurrent NSCLC (August 2018 to October 2019; 197 institutions). We evaluated plasma concentrations before the start of the third cycle of atezolizumab infusion classified into quartiles 1 to 4, their association with effectiveness, and the association between atezolizumab maximum plasma concentrations (Cmax) calculated using the existing PopPK model and AEs of special interest (AESIs). Results Overall, 175 of 262 patients were included; baseline characteristics were similar to those of patients enrolled in J-TAIL (Eastern Cooperative Oncology Group performance status ≥ 2, 12.0%; age ≥ 75 y, 28.9%; atezolizumab as more than or equal to third-line treatment, 57.5%). Atezolizumab plasma concentrations were similar to previously reported data among Japanese/non-Japanese patients. The overall survival was significantly shorter in patients with lower atezolizumab plasma concentrations in Q1 versus Q2 to Q4, although progression-free survival remained the same. The PK data adequately fit the PopPK model, with the frequency of AESIs increasing as the calculated Cmax at cycle 1 increased. Conclusions In real-world Japanese patients with unresectable advanced/recurrent NSCLC, PKs were similar to previous reports. Certain patient populations had shorter overall survival, and atezolizumab plasma concentrations in cycle 3 were lower in this population. Elevated Cmax at cycle 1 may be associated with an increased frequency of AESIs.
Collapse
Affiliation(s)
- Shigehiro Yagishita
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasushi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroaki Akamatsu
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Faculty of Medicine, Kindai University, Osaka, Japan
| | - Satoru Miura
- Department of Internal Medicine, Niigata Cancer Center Hospital, Niigata, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroshi Kagamu
- Department of Respiratory Medicine, International Medical Center, Saitama Medical University, Saitama, Japan
| | - Akinobu Hamada
- Division of Molecular Pharmacology, National Cancer Center Research Institute, Tokyo, Japan
| | - Mayu Ohuchi
- Department of Pharmacology and Therapeutics, Fundamental Innovative Oncology Core, National Cancer Center Research Institute, Tokyo, Japan
| | - Akihiko Gemma
- Department of Pulmonary Medicine and Oncology, Nippon Medical School, Graduate School of Medicine, Tokyo, Japan
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
- Department of Thoracic Surgery, International University of Health and Welfare Narita Hospital, Narita, Japan
| | - Toshihiro Misumi
- Department of Biostatistics, Yokohama City University School of Medicine, Kanagawa, Japan
- Department of Data Science, National Cancer Center Hospital East, Chiba, Japan
| | - Akito Hata
- Department of Thoracic Oncology, Kobe Minimally Invasive Cancer Center, Hyogo, Japan
| | - Satoshi Hara
- Department of Respiratory Medicine, Itami City Hospital, Hyogo, Japan
| | - Takashi Kijima
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Fujita Masaki
- Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | | | | | | | - Tetsuya Mitsudomi
- Kindai Hospital Global Research Alliance Center and Thoracic Surgery, Kindai University, Osaka, Japan
| |
Collapse
|
14
|
Ji Y, Sy SKB. Utility and impact of quantitative pharmacology on dose selection and clinical development of immuno-oncology therapy. Cancer Chemother Pharmacol 2024; 93:273-293. [PMID: 38430307 DOI: 10.1007/s00280-024-04643-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/23/2024] [Indexed: 03/03/2024]
Abstract
Immuno-oncology (IO) therapies have changed the cancer treatment landscape. Immune checkpoint inhibitors (ICIs) have improved overall survival in 20-40% of patients with malignancies that were previously refractory. Due to the uniqueness in biology, modalities and patient responses, drug development strategies for IO differed from that traditionally used for cytotoxic and target therapies in oncology, and quantitative pharmacology utilizing modeling approach can be applied in all phases of the development process. In this review, we used case studies to showcase how various modeling methodologies were applied from translational science and dose selection through to label change, using examples that included anti-programmed-death-1 (anti-PD-1), anti-programmed-death ligand-1 (anti-PD-L1), anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4), and anti-glucocorticoid-induced tumor necrosis factor receptor-related protein (anti-GITR) antibodies. How these approaches were utilized to support phase I-III dose selection, the design of phase III trials, and regulatory decisions on label change are discussed to illustrate development strategies. Model-based quantitative approaches have positively impacted IO drug development, and a better understanding of the biology and exposure-response relationship may benefit the development and optimization of new IO therapies.
Collapse
Affiliation(s)
- Yan Ji
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, 07936, USA.
| | - Sherwin K B Sy
- Novartis Pharmaceuticals Corporation, 1 Health Plaza, East Hanover, NJ, 07936, USA.
| |
Collapse
|
15
|
Bergsma EJ, Elgawly M, Mancuso D, Orr R, Vuskovich T, Seligson ND. Atezolizumab as the First Systemic Therapy Approved for Alveolar Soft Part Sarcoma. Ann Pharmacother 2024; 58:407-415. [PMID: 37466080 DOI: 10.1177/10600280231187421] [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: 07/20/2023] Open
Abstract
OBJECTIVE The objective was to review the pharmacology, efficacy, and safety of atezolizumab (Tecentriq) for the treatment of adult and pediatric patients aged 2 years and older with unresectable or metastatic alveolar soft part sarcoma (ASPS). DATA SOURCES A literature search was conducted using PubMed and MEDLINE databases, published abstracts, and ongoing studies from ClinicalTrials.gov between January 1, 1981, and May 31, 2023. Keywords included atezolizumab, Tecentriq, MPDL3280, immunotherapy, PD-L1, PD-1, pediatrics, sarcoma, and ASPS. STUDY SELECTION AND DATA EXTRACTION All English-language studies involving atezolizumab for ASPS were included and discussed. DATA SYNTHESIS Atezolizumab is an anti-programmed death-ligand 1 (PD-L1) monoclonal antibody designed to block the interaction between PD-L1 and the programmed cell death protein 1 (PD-1) receptor. Atezolizumab was granted approval by the FDA specifically for ASPS based on a phase II clinical trial in adult and pediatric patients (n = 49), which reported an overall response rate of 24% and a durable response rate at 6 and 12 months of 67% and 42%, respectively. Common grade 3/4 adverse reactions include musculoskeletal pain (8%), followed by hypertension (6%), weight gain (6%), headache (4%), and dizziness (4%). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IN COMPARISON WITH EXISTING DRUGS Advanced ASPS is a high-risk disease with limited treatment options. Atezolizumab appears to be a viable treatment option in ASPS demonstrating clinical efficacy and a manageable toxicity profile. CONCLUSIONS With no other treatments that are FDA approved specifically for ASPS, and few demonstrating efficacy in the advanced setting, the approval of atezolizumab, including the first approval for pediatric patients, represents a landmark improvement to the therapeutic arsenal against this rare disease.
Collapse
Affiliation(s)
- Emilie J Bergsma
- Department of Pharmacy, University of Florida Health Shands Hospital, Gainesville, FL, USA
| | - Mariam Elgawly
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - David Mancuso
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Roger Orr
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Theresa Vuskovich
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
| | - Nathan D Seligson
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Jacksonville, FL, USA
- Precision Medicine, Nemours Children's Health, Jacksonville, FL, USA
| |
Collapse
|
16
|
Haq ATA, Yang PP, Jin C, Shih JH, Chen LM, Tseng HY, Chen YA, Weng YS, Wang LH, Snyder MP, Hsu HL. Immunotherapeutic IL-6R and targeting the MCT-1/IL-6/CXCL7/PD-L1 circuit prevent relapse and metastasis of triple-negative breast cancer. Theranostics 2024; 14:2167-2189. [PMID: 38505617 PMCID: PMC10945351 DOI: 10.7150/thno.92922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/22/2024] [Indexed: 03/21/2024] Open
Abstract
Rationale: Multiple copies in T-cell malignancy 1 (MCT-1) is a prognostic biomarker for aggressive breast cancers. Overexpressed MCT-1 stimulates the IL-6/IL-6R/gp130/STAT3 axis, which promotes epithelial-to-mesenchymal transition and cancer stemness. Because cancer stemness largely contributes to the tumor metastasis and recurrence, we aimed to identify whether the blockade of MCT-1 and IL-6R can render these effects and to understand the underlying mechanisms that govern the process. Methods: We assessed primary tumor invasion, postsurgical local recurrence and distant metastasis in orthotopic syngeneic mice given the indicated immunotherapy and MCT-1 silencing (shMCT-1). Results: We found that shMCT-1 suppresses the transcriptomes of the inflammatory response and metastatic signaling in TNBC cells and inhibits tumor recurrence, metastasis and mortality in xenograft mice. IL-6R immunotherapy and shMCT-1 combined further decreased intratumoral M2 macrophages and T regulatory cells (Tregs) and avoided postsurgical TNBC expansion. shMCT-1 also enhances IL-6R-based immunotherapy effectively in preventing postsurgical TNBC metastasis, recurrence and mortality. Anti-IL-6R improved helper T, cytotoxic T and natural killer (NK) cells in the lymphatic system and decreased Tregs in the recurrent and metastatic tumors. Combined IL-6R and PD-L1 immunotherapies abridged TNBC cell stemness and M2 macrophage activity to a greater extent than monotherapy. Sequential immunotherapy of PD-L1 and IL-6R demonstrated the best survival outcome and lowest postoperative recurrence and metastasis compared with synchronized therapy, particularly in the shMCT-1 context. Multiple positive feedforward loops of the MCT-1/IL-6/IL-6R/CXCL7/PD-L1 axis were identified in TNBC cells, which boosted metastatic niches and immunosuppressive microenvironments. Clinically, MCT-1high/PD-L1high/CXCL7high and CXCL7high/IL-6high/IL-6Rhigh expression patterns predict worse prognosis and poorer survival of breast cancer patients. Conclusion: Systemic targeting the MCT-1/IL-6/IL-6R/CXCL7/PD-L1 interconnections enhances immune surveillance that inhibits the aggressiveness of TNBC.
Collapse
Affiliation(s)
- Aushia Tanzih Al Haq
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Pao-Pao Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Christopher Jin
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jou-Ho Shih
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Li-Mei Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Hong-Yu Tseng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Yen-An Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Yueh-Shan Weng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Lu-Hai Wang
- Institute of Integrated Medicine and Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hsin-Ling Hsu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
17
|
Ait-Oudhia S, Wang YM, Dosne AG, Roy A, Jin JY, Shen J, Kagan L, Musuamba FT, Zhang L, Kijima S, Gastonguay MR, Ouellet D. Challenging the Norm: A Multidisciplinary Perspective on Intravenous to Subcutaneous Bridging Strategies for Biologics. Clin Pharmacol Ther 2024; 115:412-421. [PMID: 38069528 DOI: 10.1002/cpt.3133] [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: 09/22/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023]
Abstract
The transition from intravenous (i.v.) to subcutaneous (s.c.) administration of biologics is a critical strategy in drug development aimed at improving patient convenience, compliance, and therapeutic outcomes. Focusing on the increasing role of model-informed drug development (MIDD) in the acceleration of this transition, an in-depth overview of the essential clinical pharmacology, and regulatory considerations for successful i.v. to s.c. bridging for biologics after the i.v. formulation has been approved are presented. Considerations encompass multiple aspects beginning with adequate pharmacokinetic (PK) and pharmacodynamic (i.e., exposure-response) evaluations which play a vital role in establishing comparability between the i.v. and s.c. routes of administrations. Selected key recommendations and points to consider include: (i) PK characterization of the s.c. formulation, supported by the increasing preclinical understanding of the s.c. absorption, and robust PK study design and analyses in humans; (ii) a thorough characterization of the exposure-response profiles including important metrics of exposure for both efficacy and safety; (iii) comparability studies designed to meet regulatory considerations and support approval of the s.c. formulation, including noninferiority studies with PK and/or efficacy and safety as primary end points; and (iv) comprehensive safety package addressing assessments of immunogenicity and patients' safety profile with the new route of administration. Recommendations for successful bridging strategies are evolving and MIDD approaches have been used successfully to accelerate the transition to s.c. dosing, ultimately leading to improved patient experiences, adherence, and clinical outcomes.
Collapse
Affiliation(s)
| | - Yow-Ming Wang
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anne-Gaelle Dosne
- Janssen Research & Development, LLC, Beerse, Belgium
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amit Roy
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jin Y Jin
- Genentech Inc., South San Francisco, California, USA
| | - Jun Shen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Leonid Kagan
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Flora T Musuamba
- Belgian Federal Agency for Medicines and Health Products, Brussels, Belgium
- NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Lucia Zhang
- Health Canada, Biologic and Radiopharmaceutical Drugs Directorate, Ottawa, Ontario, Canada
| | - Shinichi Kijima
- Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan
| | | | - Daniele Ouellet
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
18
|
Bokemeyer C, Ciardiello F, Dubreuil O, Guigay J, Kasper S, Pfeiffer P, Pinto C, Yamaguchi K, Yoshino T, Zielinski C, Esser R, Tabernero J. Cetuximab every 2 weeks versus standard weekly dosing administration schedule. Future Oncol 2024; 20:393-407. [PMID: 37850363 DOI: 10.2217/fon-2023-0282] [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] [Indexed: 10/19/2023] Open
Abstract
Cetuximab every 2 weeks (Q2W) dosing schedule is approved by the US FDA and by the Japanese Pharmaceuticals and Medical Devices Agency in patients with metastatic colorectal cancer and squamous cell carcinoma of the head and neck. Phase II trials have found comparable efficacy and safety for the weekly (Q1W) and Q2W schedules, and real-world studies have shown noninferiority of the Q2W compared with the Q1W schedule. Several guidelines recommend cetuximab Q2W administration as an alternative to the Q1W dosing schedule. Cetuximab Q2W can be administered with a Q2W dose of chemotherapy, making it a more convenient option to the Q1W schedule, potentially resulting in reduced costs for administration, increased flexibility for clinical staff and improved patient adherence.
Collapse
Affiliation(s)
- Carsten Bokemeyer
- The II Medical Clinic, Department of Oncology, Hematology & BMT with section of Pneumology, University Medical Center of Hamburg-Eppendorf, Hamburg, Germany
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Olivier Dubreuil
- Medical Oncology Unit, Diaconesses-Croix St Simon Hospital, Paris, France
| | - Joel Guigay
- Groupe d'Oncologie Radiothérapie Tête Et Cou (GORTEC), Tours, France
| | - Stefan Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Carmine Pinto
- Medical Oncology, Comprehensive Cancer Center, AUSL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Kensei Yamaguchi
- Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology & Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | | | | | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus & Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| |
Collapse
|
19
|
Drakaki A, Powles T, Bamias A, Martin-Liberal J, Shin SJ, Friedlander T, Tosi D, Park C, Gomez-Roca C, Joly Lobbedez F, Castellano D, Morales-Barrera R, Moreno-Candilejo I, Fléchon A, Yuen K, Rishipathak D, DuPree K, Young F, Michielin F, Shemesh CS, Steinberg EE, Williams P, Lee JL. Atezolizumab plus Magrolimab, Niraparib, or Tocilizumab versus Atezolizumab Monotherapy in Platinum-Refractory Metastatic Urothelial Carcinoma: A Phase Ib/II Open-Label, Multicenter, Randomized Umbrella Study (MORPHEUS Urothelial Carcinoma). Clin Cancer Res 2023; 29:4373-4384. [PMID: 37651261 DOI: 10.1158/1078-0432.ccr-23-0798] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/27/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
PURPOSE The MORPHEUS platform was designed to identify early efficacy signals and evaluate the safety of novel immunotherapy combinations across cancer types. The phase Ib/II MORPHEUS-UC trial (NCT03869190) is evaluating atezolizumab plus magrolimab, niraparib, or tocilizumab in platinum-refractory locally advanced or metastatic urothelial carcinoma (mUC). Additional treatment combinations were evaluated and will be reported separately. PATIENTS AND METHODS Patients had locally advanced or mUC that progressed during or following treatment with a platinum-containing regimen. The primary efficacy endpoint was investigator-assessed objective response rate (ORR). Key secondary endpoints included investigator-assessed progression-free survival (PFS) and overall survival (OS). Safety and exploratory biomarker analyses were also conducted. RESULTS Seventy-six patients were randomized to receive either atezolizumab plus magrolimab (n = 16), atezolizumab plus niraparib (n = 15), atezolizumab plus tocilizumab (n = 15), or atezolizumab monotherapy (control; n = 30). No additive benefit in ORR, PFS, or OS was seen in the treatment arms versus the control. The best confirmed ORR was 26.7% with atezolizumab plus magrolimab, 6.7% with atezolizumab plus niraparib, 20.0% with atezolizumab plus tocilizumab, and 27.6% with atezolizumab monotherapy. Overall, the treatment combinations were tolerable, and adverse events were consistent with each agent's known safety profile. Trends were observed for shrinkage of programmed death-ligand 1-positive tumors (atezolizumab, atezolizumab plus magrolimab, atezolizumab plus tocilizumab), inflamed tumors, or tumors with high mutational burden (atezolizumab), and immune excluded tumors (atezolizumab plus magrolimab). CONCLUSIONS The evaluated regimens in MORPHEUS-UC were tolerable. However, response rates for the combinations did not meet the criteria for further development in platinum-experienced locally advanced or mUC.
Collapse
Affiliation(s)
- Alexandra Drakaki
- Division of Hematology and Oncology, UCLA David Geffen School of Medicine, Los Angeles, California
| | - Thomas Powles
- Barts Cancer Centre, Barts Health NHS Trust, Queen Mary University of London, London, United Kingdom
| | | | - Juan Martin-Liberal
- Medical Oncology Department, Catalan Institute of Oncology (ICO) Hospitalet, Barcelona, Spain
| | - Sang Joon Shin
- Yonsei Cancer Center, Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Terence Friedlander
- University of California San Francisco, Helen Diller Family Cancer Center, San Francisco, California
| | - Diego Tosi
- Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Carlos Gomez-Roca
- Department of Medical Oncology, Institut Claudius Regaud/IUCT Oncopole, Toulouse, France
| | | | | | - Rafael Morales-Barrera
- Vall d'Hebron Institute of Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | - Kobe Yuen
- Genentech, Inc., South San Francisco, California
| | | | - Kelly DuPree
- Genentech, Inc., South San Francisco, California
| | - Fiona Young
- Roche Products Ltd, Welwyn Garden City, United Kingdom
| | | | | | | | | | - Jae Lyun Lee
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
20
|
Marolleau S, Mogenet A, Boeri C, Hamimed M, Ciccolini J, Greillier L. Killing a fly with a sledgehammer: Atezolizumab exposure in real-world lung cancer patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:1795-1803. [PMID: 38011601 PMCID: PMC10681534 DOI: 10.1002/psp4.13063] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 11/29/2023] Open
Abstract
Atezolizumab is an anti-PDL1 approved for treating lung cancer. A threshold of 6 μg/mL in plasma has been associated with target engagement. The extent to which patients could be overexposed with the standard 1200 mg q3w dosing remains unknown. Here, we monitored atezolizumab peak and trough levels in 27 real-world patients with lung cancer as part of routine therapeutic drug monitoring. Individual pharmacokinetic (PK) parameters were calculated using a population approach and optimal dosing-intervals were simulated with respect to the target trough levels. No patient had plasma levels below 6 μg/mL. The results showed that the mean trough level after the first treatment was 78.3 ± 17 μg/mL, that is, 13 times above the target concentration. The overall response rate was 55.5%. Low-grade immune-related adverse events was observed in 37% of patients. No relationship was found between exposure metrics of atezolizumab (i.e., minimum plasma concentration, maximum plasma concentration, and area under the curve) and pharmacodynamic end points (i.e., efficacy and toxicity). Further simulations suggest that the dosing interval could be extended from 21 days to 49 up to 136 days (mean: 85.7 days, i.e., q12w), while ensuring plasma levels still above the 6 μg/mL target threshold. This observational, real-world study suggests that the standard 1200 mg q3w fixed-dose regimen of atezolizumab results in significant overexposure in all the patients. This was not associated with increased side effects. As plasma levels largely exceed pharmacologically active concentrations, interindividual variability in PK parameters did not impact efficacy. Our data suggest that dosing intervals could be markedly extended with respect to the target threshold associated with efficacy.
Collapse
Affiliation(s)
- Sophie Marolleau
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Alice Mogenet
- Oncologie multidisciplinaire et innovations thérapeutiquesNord University Hospital of MarseilleMarseilleFrance
| | - Clara Boeri
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Mourad Hamimed
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Joseph Ciccolini
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
| | - Laurent Greillier
- COMPO, Inserm U1068 Centre de Recherche en Cancérologie de Marseille & Inria Sophia AntipolisMarseilleFrance
- Oncologie multidisciplinaire et innovations thérapeutiquesNord University Hospital of MarseilleMarseilleFrance
| |
Collapse
|
21
|
Ratain MJ, Strohbehn GW. Combining atezolizumab 1200 mg with bevacizumab 15 mg/kg: based on science or just revenues? Eur J Cancer 2023; 194:113349. [PMID: 37806256 DOI: 10.1016/j.ejca.2023.113349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023]
Abstract
Dose optimisation is increasingly important in oncology, as exemplified by the US Food and Drug Administration's Project Optimus initiative, which is aligned with similar initiatives in other countries. In parallel, multiple stakeholders have raised concerns about anticancer drug prices, affordability, and access. This is of particular concern to government payers as well as patients and physicians in low- and middle-income countries. As anticancer drugs have historically been approved at the maximally tolerated dose, it is now highly relevant to question whether lower doses are equally effective and can be delivered at lower doses, resulting in less toxicity for patients, and lower costs for patients and payers. We illustrate this opportunity by discussing the combination of atezolizumab and bevacizumab, approved in multiple countries for both non-small cell lung cancer and hepatocellular cancer. Our conclusion is that the cost of this regimen can be reduced by more than 80%, an opportunity that should be considered by patients, prescribers, payers, and policymakers.
Collapse
Affiliation(s)
- Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA.
| | - Garth W Strohbehn
- Veterans Affairs Center for Clinical Management Research and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
22
|
Strobl MAR, Gallaher J, Robertson-Tessi M, West J, Anderson ARA. Treatment of evolving cancers will require dynamic decision support. Ann Oncol 2023; 34:867-884. [PMID: 37777307 PMCID: PMC10688269 DOI: 10.1016/j.annonc.2023.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 10/02/2023] Open
Abstract
Cancer research has traditionally focused on developing new agents, but an underexplored question is that of the dose and frequency of existing drugs. Based on the modus operandi established in the early days of chemotherapies, most drugs are administered according to predetermined schedules that seek to deliver the maximum tolerated dose and are only adjusted for toxicity. However, we believe that the complex, evolving nature of cancer requires a more dynamic and personalized approach. Chronicling the milestones of the field, we show that the impact of schedule choice crucially depends on processes driving treatment response and failure. As such, cancer heterogeneity and evolution dictate that a one-size-fits-all solution is unlikely-instead, each patient should be mapped to the strategy that best matches their current disease characteristics and treatment objectives (i.e. their 'tumorscape'). To achieve this level of personalization, we need mathematical modeling. In this perspective, we propose a five-step 'Adaptive Dosing Adjusted for Personalized Tumorscapes (ADAPT)' paradigm to integrate data and understanding across scales and derive dynamic and personalized schedules. We conclude with promising examples of model-guided schedule personalization and a call to action to address key outstanding challenges surrounding data collection, model development, and integration.
Collapse
Affiliation(s)
- M A R Strobl
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa; Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - J Gallaher
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - M Robertson-Tessi
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - J West
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - A R A Anderson
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa.
| |
Collapse
|
23
|
Choudhary N, Bawari S, Burcher JT, Sinha D, Tewari D, Bishayee A. Targeting Cell Signaling Pathways in Lung Cancer by Bioactive Phytocompounds. Cancers (Basel) 2023; 15:3980. [PMID: 37568796 PMCID: PMC10417502 DOI: 10.3390/cancers15153980] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is a heterogeneous group of malignancies with high incidence worldwide. It is the most frequently occurring cancer in men and the second most common in women. Due to its frequent diagnosis and variable response to treatment, lung cancer was reported as the top cause of cancer-related deaths worldwide in 2020. Many aberrant signaling cascades are implicated in the pathogenesis of lung cancer, including those involved in apoptosis (B cell lymphoma protein, Bcl-2-associated X protein, first apoptosis signal ligand), growth inhibition (tumor suppressor protein or gene and serine/threonine kinase 11), and growth promotion (epidermal growth factor receptor/proto-oncogenes/phosphatidylinositol-3 kinase). Accordingly, these pathways and their signaling molecules have become promising targets for chemopreventive and chemotherapeutic agents. Recent research provides compelling evidence for the use of plant-based compounds, known collectively as phytochemicals, as anticancer agents. This review discusses major contributing signaling pathways involved in the pathophysiology of lung cancer, as well as currently available treatments and prospective drug candidates. The anticancer potential of naturally occurring bioactive compounds in the context of lung cancer is also discussed, with critical analysis of their mechanistic actions presented by preclinical and clinical studies.
Collapse
Affiliation(s)
- Neeraj Choudhary
- Department of Pharmacognosy, GNA School of Pharmacy, GNA University, Phagwara 144 401, India
| | - Sweta Bawari
- Amity Institute of Pharmacy, Amity University, Noida 201 301, India
| | - Jack T. Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110 017, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| |
Collapse
|
24
|
Burotto M, Zvirbule Z, Mochalova A, Runglodvatana Y, Herraez-Baranda L, Liu SN, Chan P, Shearer-Kang E, Liu X, Tosti N, Zanghi JA, Leutgeb B, Felip E. IMscin001 Part 2: a randomised phase III, open-label, multicentre study examining the pharmacokinetics, efficacy, immunogenicity, and safety of atezolizumab subcutaneous versus intravenous administration in previously treated locally advanced or metastatic non-small-cell lung cancer and pharmacokinetics comparison with other approved indications. Ann Oncol 2023; 34:693-702. [PMID: 37268157 DOI: 10.1016/j.annonc.2023.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 04/19/2023] [Accepted: 05/22/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Atezolizumab intravenous (IV) is approved for the treatment of various solid tumours. To improve treatment convenience and health care efficiencies, a coformulation of atezolizumab and recombinant human hyaluronidase PH20 was developed for subcutaneous (SC) use. Part 2 of IMscin001 (NCT03735121) was a randomised phase III, open-label, multicentre, noninferiority study comparing the drug exposure of atezolizumab SC with atezolizumab IV. PATIENTS AND METHODS Eligible patients with locally advanced/metastatic non-small-cell lung cancer were randomised 2 : 1 to receive atezolizumab SC (1875 mg; n = 247) or IV (1200 mg; n = 124) every 3 weeks. The co-primary endpoints were cycle 1 observed trough serum concentration (Ctrough) and model-predicted area under the curve from days 0 to 21 (AUC0-21 d). The secondary endpoints were steady-state exposure, efficacy, safety, and immunogenicity. Exposure following atezolizumab SC was then compared with historical atezolizumab IV values across approved indications. RESULTS The study met both of its co-primary endpoints: cycle 1 observed Ctrough {SC: 89 μg/ml [coefficient of variation (CV): 43%] versus IV: 85 μg/ml (CV: 33%); geometric mean ratio (GMR), 1.05 [90% confidence interval (CI) 0.88-1.24]} and model-predicted AUC0-21 d [SC: 2907 μg d/ml (CV: 32%) versus IV: 3328 μg d/ml (CV: 20%); GMR, 0.87 (90% CI 0.83-0.92)]. Progression-free survival [hazard ratio 1.08 (95% CI 0.82-1.41)], objective response rate (SC: 12% versus IV: 10%), and incidence of anti-atezolizumab antibodies (SC: 19.5% versus IV: 13.9%) were similar between arms. No new safety concerns were identified. Ctrough and AUC0-21 d for atezolizumab SC were consistent with the other approved atezolizumab IV indications. CONCLUSIONS Compared with IV, atezolizumab SC demonstrated noninferior drug exposure at cycle 1. Efficacy, safety, and immunogenicity were similar between arms and consistent with the known profile for atezolizumab IV. Similar drug exposure and clinical outcomes following SC and IV administration support the use of atezolizumab SC as an alternative to atezolizumab IV.
Collapse
Affiliation(s)
- M Burotto
- Centro de Investigación, Clínica Bradford Hill, Santiago, Chile.
| | - Z Zvirbule
- Latvian Oncology Center, Riga Eastern Clinical University Hospital, Riga, Latvia
| | - A Mochalova
- Department of Antitumor Drug Therapy, MEDSI Clinical Hospital, Moscow, Russia
| | - Y Runglodvatana
- Faculty of Medicine, Vajira Hospital, Bangkok Metropolitan University, Bangkok, Thailand
| | - L Herraez-Baranda
- Product Development Medical Affairs Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - S N Liu
- Clinical Pharmacology, Genentech, Inc., South San Francisco, USA
| | - P Chan
- Clinical Pharmacology, Genentech, Inc., South San Francisco, USA
| | - E Shearer-Kang
- Product Safety Development, Genentech, Inc., South San Francisco, CA, USA
| | - X Liu
- Data and Statistical Sciences, Genentech, Inc., South San Francisco, CA, USA
| | - N Tosti
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - J A Zanghi
- BioAnalytical Sciences, Genentech, Inc., South San Francisco, CA, USA
| | - B Leutgeb
- Product Development Oncology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - E Felip
- Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain; Clinical Research Department, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
25
|
Endo Y, Winarski KL, Sajib MS, Ju A, Wu WJ. Atezolizumab Induces Necroptosis and Contributes to Hepatotoxicity of Human Hepatocytes. Int J Mol Sci 2023; 24:11694. [PMID: 37511454 PMCID: PMC10380327 DOI: 10.3390/ijms241411694] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/13/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Atezolizumab is an immune checkpoint inhibitor (ICI) targeting PD-L1 for treatment of solid malignancies. Immune checkpoints control the immune tolerance, and the adverse events such as hepatotoxicity induced by ICIs are often considered as an immune-related adverse event (irAE). However, PD-L1 is also highly expressed in normal tissues, e.g., hepatocytes. It is still not clear whether, targeting PD-L1 on hepatocytes, the atezolizumab may cause damage to liver cells contributing to hepatotoxicity. Here, we reveal a novel mechanism by which the atezolizumab induces hepatotoxicity in human hepatocytes. We find that the atezolizumab treatment increases a release of LDH in the cell culture medium of human hepatocytes (human primary hepatocytes and THLE-2 cells), decreases cell viability, and inhibits the THLE-2 and THLE-3 cell growth. We demonstrate that both the atezolizumab and the conditioned medium (T-CM) derived from activated T cells can induce necroptosis of the THLE-2 cells, which is underscored by the fact that the atezolizumab and T-CM enhance the phosphorylation of RIP3 and MLKL proteins. Furthermore, we also show that necrostatin-1, a necrosome inhibitor, decreases the amount of phosphorylated RIP3 induced by the atezolizumab, resulting in a reduced LDH release in the culture media of the THLE-2 cells. This finding is further supported by the data that GSK872 (a RIP3 inhibitor) significantly reduced the atezolizumab-induced LDH release. Taken together, our data indicate that the atezolizumab induces PD-L1-mediated necrosome formation, contributing to hepatotoxicity in PD-L1+-human hepatocytes. This study provides the molecular basis of the atezolizumab-induced hepatotoxicity and opens a new avenue for developing a novel therapeutic approach to reducing hepatotoxicity induced by ICIs.
Collapse
Affiliation(s)
- Yukinori Endo
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Katie L Winarski
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Md Sanaullah Sajib
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Anna Ju
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| | - Wen Jin Wu
- Division of Biotechnology Review and Research 1, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration (FDA), Silver Spring, MD 20993, USA
| |
Collapse
|
26
|
Barrett JS, Goyal RK, Gobburu J, Baran S, Varshney J. An AI Approach to Generating MIDD Assets Across the Drug Development Continuum. AAPS J 2023; 25:70. [PMID: 37430126 DOI: 10.1208/s12248-023-00838-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023] Open
Abstract
Model-informed drug development involves developing and applying exposure-based, biological, and statistical models derived from preclinical and clinical data sources to inform drug development and decision-making. Discrete models are generated from individual experiments resulting in a single model expression that is utilized to inform a single stage-gate decision. Other model types provide a more holistic view of disease biology and potentially disease progression depending on the appropriateness of the underlying data sources for that purpose. Despite this awareness, most data integration and model development approaches are still reliant on internal (within company) data stores and traditional structural model types. An AI/ML-based MIDD approach relies on more diverse data and is informed by past successes and failures including data outside a host company (external data sources) that may enhance predictive value and enhance data generated by the sponsor to reflect more informed and timely experimentation. The AI/ML methodology also provides a complementary approach to more traditional modeling efforts that support MIDD and thus yields greater fidelity in decision-making. Early pilot studies support this assessment but will require broader adoption and regulatory support for more evidence and refinement of this paradigm. An AI/ML-based approach to MIDD has the potential to transform regulatory science and the current drug development paradigm, optimize information value, and increase candidate and eventually product confidence with respect to safety and efficacy. We highlight early experiences with this approach using the AI compute platforms as representative examples of how MIDD can be facilitated with an AI/ML approach.
Collapse
Affiliation(s)
- Jeffrey S Barrett
- Aridhia Bioinformatics, 163 Bath Street, Glasgow, Scotland, G2 4SQ, UK.
| | - Rahul K Goyal
- Center for Translational Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA
| | - Jogarao Gobburu
- Center for Translational Medicine, University of Maryland Baltimore, Baltimore, Maryland, USA
- Pumas-AI, Baltimore, Maryland, USA
| | | | | |
Collapse
|
27
|
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
|
28
|
Peer CJ, Schmidt KT, Arisa O, Richardson WJ, Paydary K, Goldstein DA, Gulley JL, Figg WD, Ratain MJ. In Silico Re-Optimization of Atezolizumab Dosing Using Population Pharmacokinetic Simulation and Exposure-Response Simulation. J Clin Pharmacol 2023; 63:672-680. [PMID: 36624662 PMCID: PMC10175103 DOI: 10.1002/jcph.2203] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
Atezolizumab, a humanized monoclonal antibody against programmed cell death ligand 1 (PD-L1), was initially approved in 2016, around the same time that the sponsor published the minimum serum concentration to maintain the saturation of receptor occupancy (6 μg/mL). The initially approved dose regimen of 1200 mg every 3 weeks (q3w) was subsequently modified to 840 mg q2w or 1680 mg q4w through pharmacokinetic simulations. Yet, each standard regimen yields steady-state trough concentrations (CMIN,SS ) far exceeding (≈ 40-fold) the stated target concentration. Additionally, the steady-state area under the plasma drug concentration-time curve (AUCSS ) at 1200 mg q3w was significantly (P = .027) correlated with the probability of adverse events of special interest (AESIs) in patients with non-small cell lung cancer (NSCLC) and, coupled with excess exposure, this provides incentive to explore alternative dose regimens to lower the exposure burden while maintaining an effective CMIN,SS . In this study, we first identified 840 mg q6w as an extended-interval regimen that could robustly maintain a serum concentration of 6 μg/mL (≥99% of virtual patients simulated, n = 1000), then applied this regimen to an approach that administers 2 "loading doses" of standard-interval regimens for a future clinical trial aiming to personalize dose regimens. Each standard dose was simulated for 2 loading doses, then 840 mg q6w thereafter; all yielded cycle-7 CMIN,SS values of >6 μg/mL in >99% of virtual patients. Further, the AUCSS from 840 mg q6w resulted in a flattening (P = .63) of the exposure-response relationship with adverse events of special interest (AESIs). We next aim to verify this in a clinical trial seeking to validate extended-interval dosing in a personalized approach using therapeutic drug monitoring.
Collapse
Affiliation(s)
- Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Keith T. Schmidt
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Oluwatobi Arisa
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | | | - Koosha Paydary
- Department of Medicine, Section of Hematology/Oncology, and Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| | - Daniel A. Goldstein
- Faculty of Medicine, Tel Aviv University, Israel
- Davidoff Cancer Center, Rabin Medical Center, Israel
- Clalit Health Services, Tela Aviv, Israel
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, MD, USA
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Mark J. Ratain
- Department of Medicine, Section of Hematology/Oncology, and Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
29
|
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
|
30
|
Soltantabar P, Lon HK, Parivar K, Wang DD, Elmeliegy M. Optimizing benefit/risk in oncology: Review of post-marketing dose optimization and reflections on the road ahead. Crit Rev Oncol Hematol 2023; 182:103913. [PMID: 36681205 DOI: 10.1016/j.critrevonc.2023.103913] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Oncology therapies shifted from chemotherapy to molecularly targeted agents and finally to the era of immune-oncology agents. In contrast to cytotoxic agents, molecularly targeted agents are more selective, exhibit a wider therapeutic window, and may maximally modulate tumor growth at doses lower than the maximum tolerated dose (MTD). However, first-in-patient oncology studies for molecularly targeted agents continued to evaluate escalating doses using limited number of patients per dose cohort assessing dose-limiting toxicities to identify the MTD which is commonly selected for further development adopting a 'more is better' approach that led to several post-marketing requirement (PMR) studies to evaluate alternative, typically lower, doses or dosing frequencies to optimize the benefit-risk profile. In this review, post-marketing dose optimization efforts were reviewed including those required by a regulatory pathway or voluntarily conducted by the sponsor to improve efficacy, safety, or method of administration. Lessons learned and future implications from this deep dive review are discussed considering the evolving regulatory landscape on dose optimization for oncology compounds.
Collapse
Affiliation(s)
| | - Hoi-Kei Lon
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | | - Diane D Wang
- Global Product Development, Pfizer Inc, San Diego, CA, USA
| | | |
Collapse
|
31
|
Overbeek JK, Ter Heine R, Verheul HMW, Chatelut E, Rudek MA, Gurney H, Plummer R, Gilbert DC, Buclin T, Burger DM, Bloemendal HJ, van Erp NP. Off-label, but on target: the evidence needed to implement alternative dosing regimens of anticancer drugs. ESMO Open 2023; 8:100749. [PMID: 36603522 PMCID: PMC9813708 DOI: 10.1016/j.esmoop.2022.100749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 01/05/2023] Open
Affiliation(s)
- J K Overbeek
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - R Ter Heine
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - H M W Verheul
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen; Department of Medical Oncology, Erasmus University MC Cancer Institute, Rotterdam, Netherlands
| | - E Chatelut
- Institut Claudius-Regaud, IUCT-Oncopole, and CRCT, Université de Toulouse, Inserm, 1, Toulouse, France; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore
| | - M A Rudek
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, USA; Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore; Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, USA
| | - H Gurney
- Department of Clinical Medicine, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, Australia
| | - R Plummer
- Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne
| | - D C Gilbert
- MRC Clinical Trials Unit, University College London (UCL), Institute of Clinical Trials and Methodology, London, UK; Optimal Cancer Care Alliance, Ann Arbor, USA
| | - T Buclin
- Division of Clinical Pharmacology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - D M Burger
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - H J Bloemendal
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen
| | - N P van Erp
- Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands.
| |
Collapse
|
32
|
Laktionov KK, Artamonova EV, Borisova TN, Breder VV, Bychkov IM, Vladimirova LI, Volkov NM, Ergnian SM, Zhabina AS, Kononets PV, Kuzminov AE, Levchenko EV, Malikhova OA, Marinov DT, Miller SV, Moiseenko FV, Mochal’nikova VV, Novikov SN, Pikin OV, Reutova EV, Rodionov EO, Sakaeva DD, Sarantseva KA, Semenova AI, Smolin AV, Sotnikov VM, Tuzikov SA, Turkin IN, Tyurin IE, Chkhikvadze VD, Kolbanov KI, Chernykh MV, Chernichenko AV, Fedenko AA, Filonenko EV, Nevol’skikh AA, Ivanov SA, Khailova ZV, Gevorkian TG, Butenko AV, Gil’mutdinova IR, Gridneva IV, Eremushkin MA, Zernova MA, Kasparov BS, Kovlen DV, Kondrat’eva KO, Konchugova TV, Korotkova SB, Krutov AA, Obukhova OA, Ponomarenko GN, Semiglazova TI, Stepanova AM, Khulamkhanova MM. Malignant neoplasm of the bronchi and lung: Russian clinical guidelines. JOURNAL OF MODERN ONCOLOGY 2022. [DOI: 10.26442/18151434.2022.3.201848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
удалить
Collapse
|
33
|
Mountzios G, Aggarwal C. Pembrolizumab Administration Frequency, Dose Exposure, and Toxicity: Is Switching Safe? J Thorac Oncol 2022; 17:1155-1157. [PMID: 36192075 DOI: 10.1016/j.jtho.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 11/26/2022]
Affiliation(s)
- Giannis Mountzios
- Fourth Oncology Department and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece.
| | - Charu Aggarwal
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Abramson Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
Huang W, Stader F, Chan P, Shemesh CS, Chen Y, Gill KL, Jones HM, Li L, Rossato G, Wu B, Jin JY, Chanu P. Development of a pediatric physiologically-based pharmacokinetic model to support recommended dosing of atezolizumab in children with solid tumors. Front Pharmacol 2022; 13:974423. [PMID: 36225583 PMCID: PMC9548535 DOI: 10.3389/fphar.2022.974423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Atezolizumab has been studied in multiple indications for both pediatric and adult patient populations. Generally, clinical studies enrolling pediatric patients may not collect sufficient pharmacokinetic data to characterize the drug exposure and disposition because of operational, ethical, and logistical challenges including burden to children and blood sample volume limitations. Therefore, mechanistic modeling and simulation may serve as a tool to predict and understand the drug exposure in pediatric patients. Objective: To use mechanistic physiologically-based pharmacokinetic (PBPK) modeling to predict atezolizumab exposure at a dose of 15 mg/kg (max 1,200 mg) in pediatric patients to support dose rationalization and label recommendations. Methods: A minimal mechanistic PBPK model was used which incorporated age-dependent changes in physiology and biochemistry that are related to atezolizumab disposition such as endogenous IgG concentration and lymph flow. The PBPK model was developed using both in vitro data and clinically observed data in adults and was verified across dose levels obtained from a phase I and multiple phase III studies in both pediatric patients and adults. The verified model was then used to generate PK predictions for pediatric and adult subjects ranging from 2- to 29-year-old. Results: Individualized verification in children and in adults showed that the simulated concentrations of atezolizumab were comparable (76% within two-fold and 90% within three-fold, respectively) to the observed data with no bias for either over- or under-prediction. Applying the verified model, the predicted exposure metrics including Cmin, Cmax, and AUCtau were consistent between pediatric and adult patients with a geometric mean of pediatric exposure metrics between 0.8- to 1.25-fold of the values in adults. Conclusion: The results show that a 15 mg/kg (max 1,200 mg) atezolizumab dose administered intravenously in pediatric patients provides comparable atezolizumab exposure to a dose of 1,200 mg in adults. This suggests that a dose of 15 mg/kg will provide adequate and effective atezolizumab exposure in pediatric patients from 2- to 18-year-old.
Collapse
Affiliation(s)
- Weize Huang
- Genentech Inc, South San Francisco, CA, United States
- *Correspondence: Weize Huang,
| | | | - Phyllis Chan
- Genentech Inc, South San Francisco, CA, United States
| | | | - Yuan Chen
- Genentech Inc, South San Francisco, CA, United States
| | | | | | - Linzhong Li
- Certara UK Limited, Sheffield, United Kingdom
| | | | - Benjamin Wu
- Genentech Inc, South San Francisco, CA, United States
| | - Jin Y. Jin
- Genentech Inc, South San Francisco, CA, United States
| | - Pascal Chanu
- Genentech Inc, South San Francisco, CA, United States
| |
Collapse
|
35
|
Vugmeyster Y, Grisic AM, Brockhaus B, Rueckert P, Ruisi M, Dai H, Khandelwal A. Avelumab Dose Selection for Clinical Studies in Pediatric Patients with Solid Tumors. Clin Pharmacokinet 2022; 61:985-995. [PMID: 35484319 PMCID: PMC9287219 DOI: 10.1007/s40262-022-01111-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND OBJECTIVE A phase I/II trial evaluated the safety, antitumor activity, and pharmacokinetics of avelumab (anti-PD-L1 antibody) in pediatric patients with refractory/relapsed solid tumors (NCT03451825). This study aimed to inform avelumab dose selection in pediatric populations using population pharmacokinetic modeling and simulations. METHODS Patients aged < 18 years with refractory/relapsed solid tumors enrolled in phase I received avelumab 10 or 20 mg/kg intravenously every 2 weeks. A pediatric population pharmacokinetic model was developed via the frequentist prior approach. RESULTS Pharmacokinetic parameters from 21 patients who received avelumab 10 mg/kg (n = 6) or 20 mg/kg (n = 15) were analyzed. Patients had a wide range of weights and ages (medians, 37.3 kg and 12 years). Exposures with 10-mg/kg dosing were lower vs adult dosing, particularly in patients weighing < 40 kg, whereas 20-mg/kg dosing achieved or exceeded adult exposures, irrespective of body weight. A two-compartment linear model with time-varying clearance using body weight as a covariate, with the frequentist prior approach, best described pediatric data. In this model, optimal overlap in exposure with adult data was achieved with 800 mg every 2 weeks for patients aged ≥ 12 years and weighing ≥ 40 kg, and 15 mg/kg every 2 weeks for patients aged < 12 years or weighing < 40 kg. CONCLUSIONS Based on exposure matching, the recommended doses for further avelumab studies, including combination studies, are 15 mg/kg every 2 weeks for pediatric patients aged < 12 years or weighing < 40 kg and the adult flat dose of 800 mg every 2 weeks for pediatric patients aged ≥ 12 years and weighing ≥ 40 kg. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov NCT03451825.
Collapse
Affiliation(s)
- Yulia Vugmeyster
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Ana-Marija Grisic
- Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Brigitte Brockhaus
- Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Peter Rueckert
- Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany
| | - Mary Ruisi
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Haiqing Dai
- EMD Serono Research & Development Institute, Inc. (an affiliate of Merck KGaA), Billerica, MA, USA
| | - Akash Khandelwal
- Merck Healthcare KGaA, Frankfurter Strasse 250, 64293, Darmstadt, Germany.
| |
Collapse
|
36
|
Jiang M, Hu Y, Lin G, Chen C. Dosing Regimens of Immune Checkpoint Inhibitors: Attempts at Lower Dose, Less Frequency, Shorter Course. Front Oncol 2022; 12:906251. [PMID: 35795044 PMCID: PMC9251517 DOI: 10.3389/fonc.2022.906251] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/24/2022] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a revolutionary breakthrough in the field of cancer by modulating patient's own immune system to exert anti-tumor effects. The clinical application of ICIs is still in its infancy, and their dosing regimens need to be continuously adjusted. Pharmacokinetic/pharmacodynamic studies showed a significant plateau in the exposure-response curve, with high receptor occupancy and plasma concentrations achieved at low dose levels. Coupled with concerns about drug toxicity and heavy economic costs, there has been an ongoing quest to reevaluate the current ICI dosing regimens while preserving maximum clinical efficacy. Many clinical data showed remarkable anticancer effects with ICIs at the doses far below the approved regimens, indicating the possibility of dose reduction. Our review attempts to summarize the clinical evidence for ICIs regimens with lower-dose, less-frequency, shorter-course, and provide clues for further ICIs regimen optimization.
Collapse
Affiliation(s)
| | | | | | - Chao Chen
- Department of Radiotherapy, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Provincial Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
37
|
Kasichayanula S, Mandlekar S, Shivva V, Patel M, Girish S. Evolution of Preclinical Characterization and Insights into Clinical Pharmacology of Checkpoint Inhibitors Approved for Cancer Immunotherapy. Clin Transl Sci 2022; 15:1818-1837. [PMID: 35588531 PMCID: PMC9372426 DOI: 10.1111/cts.13312] [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: 02/18/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer immunotherapy has significantly advanced the treatment paradigm in oncology, with approvals of immuno‐oncology agents for over 16 indications, many of them first line. Checkpoint inhibitors (CPIs) are recognized as an essential backbone for a successful anticancer therapy regimen. This review focuses on the US Food and Drug Administration (FDA) regulatory approvals of major CPIs and the evolution of translational advances since their first approval close to a decade ago. In addition, critical preclinical and clinical pharmacology considerations, an overview of the pharmacokinetic and dose/regimen aspects, and a discussion of the future of CPI translational and clinical pharmacology as combination therapy becomes a mainstay of industrial immunotherapy development and in clinical practice are also discussed.
Collapse
Affiliation(s)
| | | | - Vittal Shivva
- Genentech, 1 DNA Way, South San Francisco, 94080, CA
| | - Maulik Patel
- AbbVie Inc., 1000 Gateway Blvd, South San Francisco, 94080, CA
| | - Sandhya Girish
- Gilead Sciences, 310 Lakeside Drive, Foster City, 94404, CA
| |
Collapse
|
38
|
Upfront FOLFOXIRI plus bevacizumab with or without atezolizumab in the treatment of patients with metastatic colorectal cancer (AtezoTRIBE): a multicentre, open-label, randomised, controlled, phase 2 trial. Lancet Oncol 2022; 23:876-887. [DOI: 10.1016/s1470-2045(22)00274-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022]
|
39
|
Yoshida K, Chan P, Marchand M, Zhang R, Wu B, Ballinger M, Sternheim N, Jin JY, Bruno R. Tumor Growth Inhibition-Overall Survival (TGI-OS) Model for Subgroup Analysis Based on Post-Randomization Factors: Application for Anti-drug Antibody (ADA) Subgroup Analysis of Atezolizumab in the IMpower150 Study. AAPS J 2022; 24:58. [PMID: 35484442 DOI: 10.1208/s12248-022-00710-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 11/30/2022] Open
Abstract
Longitudinal changes of tumor size or tumor-associated biomarkers have been receiving growing attention as early markers of treatment benefits. Tumor growth inhibition-overall survival (TGI-OS) models represent mathematical frameworks used to establish a link from tumor size trajectory to survival outcome with the aim of predicting survival benefit with tumor data from a small number of subjects with a short follow-up time. In the present study, we applied the TGI-OS model to assess treatment benefit in the IMpower150 study for patients who exhibited development of anti-drug antibodies (ADA). Direct comparison between subgroups of the active arm [ADA positive (ADA +) and negative (ADA -) groups] to the entire control group is not appropriate, due to potential imbalances of baseline prognostic factors between ADA + and ADA - patients. Thus, the TGI-OS modeling framework was employed to adjust for differences in prognostic factors between the ADA subgroups to more accurately estimate the treatment benefits. After adjustment, the TGI-OS model predicted comparable hazard ratios (HRs) of OS between ADA + and ADA - subgroups, suggesting that the development of ADA does not have a clinically significant impact on the treatment benefit of atezolizumab.
Collapse
Affiliation(s)
- Kenta Yoshida
- Department of Clinical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | - Phyllis Chan
- Department of Clinical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | | - Rong Zhang
- Department of Clinical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Benjamin Wu
- Department of Clinical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | | - Nitzan Sternheim
- Product Development, Genentech, Inc., South San Francisco, CA, USA
| | - Jin Y Jin
- Department of Clinical Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - René Bruno
- Clinical Pharmacology, Genentech-Roche, Marseille, France
| |
Collapse
|
40
|
Holder PG, Lim SA, Huang CS, Sharma P, Dagdas YS, Bulutoglu B, Sockolosky JT. Engineering interferons and interleukins for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114112. [PMID: 35085624 DOI: 10.1016/j.addr.2022.114112] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/08/2023]
Abstract
Cytokines are a class of potent immunoregulatory proteins that are secreted in response to various stimuli and act locally to regulate many aspects of human physiology and disease. Cytokines play important roles in cancer initiation, progression, and elimination, and thus, there is a long clinical history associated with the use of recombinant cytokines to treat cancer. However, the use of cytokines as therapeutics has been limited by cytokine pleiotropy, complex biology, poor drug-like properties, and severe dose-limiting toxicities. Nevertheless, cytokines are crucial mediators of innate and adaptive antitumor immunity and have the potential to enhance immunotherapeutic approaches to treat cancer. Development of immune checkpoint inhibitors and combination immunotherapies has reinvigorated interest in cytokines as therapeutics, and a variety of engineering approaches are emerging to improve the safety and effectiveness of cytokine immunotherapy. In this review we highlight recent advances in cytokine biology and engineering for cancer immunotherapy.
Collapse
|
41
|
Maritaz C, Broutin S, Chaput N, Marabelle A, Paci A. Immune checkpoint-targeted antibodies: a room for dose and schedule optimization? J Hematol Oncol 2022; 15:6. [PMID: 35033167 PMCID: PMC8760805 DOI: 10.1186/s13045-021-01182-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
Anti-CTLA-4 and anti-PD-1/PD-L1 immune checkpoint inhibitors are therapeutic monoclonal antibodies that do not target cancer cells but are designed to reactivate or promote antitumor immunity. Dosing and scheduling of these biologics were established according to conventional drug development models, even though the determination of a maximum tolerated dose in the clinic could only be defined for anti-CTLA-4. Given the pharmacology of these monoclonal antibodies, their high interpatient pharmacokinetic variability, the actual clinical benefit as monotherapy that is observed only in a specific subset of patients, and the substantial cost of these treatments, a number of questions arise regarding the selected dose and the dosing interval. This review aims to outline the development of these immunotherapies and considers optimization options that could be used in clinical practice.
Collapse
Affiliation(s)
- Christophe Maritaz
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Sophie Broutin
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Chaput
- Laboratory for Immunomonitoring in Oncology (LIO), Faculty of Pharmacy, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France
| | - Aurélien Marabelle
- Drug Development Unit (DITEP), LRTI U1015 INSERM, Gustave Roussy, Villejuif, France
| | - Angelo Paci
- Pharmacology Department, U1030 INSERM, University Paris-Saclay, Gustave Roussy Cancer Campus, Villejuif, France.
- Pharmacokinetic Unit, Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.
| |
Collapse
|
42
|
Hamuro L, Hu Z, Passarell J, Barcomb H, Zhang J, Goldstein S, Bello A, Roy A, Zhu L. Exposure-Response Analysis to Support Nivolumab Once Every 4 Weeks Dosing in Combination with Cabozantinib in Renal Cell Carcinoma. Clin Cancer Res 2022; 28:1603-1613. [PMID: 34980597 DOI: 10.1158/1078-0432.ccr-21-3149] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/22/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE A benefit:risk assessment for a less-frequent nivolumab 480 mg Q4W + cabozantinib 40 mg QD dosing regimen was predicted using modeling and simulation of clinical trial data from nivolumab monotherapy studies and from the nivolumab 240 mg Q2W + cabozantinib 40mg QD dosing regimen, which demonstrated clinical benefit versus sunitinib in previously untreated advanced renal cell carcinoma (aRCC) in the phase III CheckMate 9ER trial (NCT03141177). EXPERIMENTAL DESIGN Multivariable Cox proportional-hazards analyses were conducted using nivolumab monotherapy data in previously treated aRCC and data from CheckMate 9ER to evaluate progression-free survival (PFS), overall survival (OS), and grade {greater than or equal to}2 immune-mediated adverse events (IMAEs). RESULTS Nivolumab 240 mg Q2W + cabozantinib versus nivolumab showed improvement in PFS (HR, 0.38; 95% CI, 0.31-0.47), OS (HR, 0.63 95% CI, 0.46-0.85), and increased risk of grade {greater than or equal to}2 IMAEs (HR, 2.19; 95% CI, 1.79-2.67). Nivolumab exposure was not a predictor of PFS/OS or grade {greater than or equal to}2 IMAEs. Lower nivolumab clearance, male sex, higher baseline bodyweight, and Karnofsky performance (100) were each associated with PFS/OS improvements. Region and IMDC poor score were negative OS predictors. Age, baseline albumin, and programmed death ligand-1 status were not significant PFS/OS predictors. Cabozantinib was a significant grade {greater than or equal to}2 IMAE predictor, driven by diarrhea and hepatic events. Model-predicted PFS/OS and grade {greater than or equal to}2 IMAE rates were similar (<2.5% difference) for nivolumab 240 mg Q2W + cabozantinib and 480 mg Q4W + cabozantinib. CONCLUSIONS Comparable benefit:risk was predicted for nivolumab 480 mg Q4W + cabozantinib and nivolumab 240 mg Q2W + cabozantinib.
Collapse
Affiliation(s)
- Lora Hamuro
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb (United States)
| | - Zheyi Hu
- Bristol-Myers Squibb (United States)
| | | | | | | | | | - Akintunde Bello
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb (United States)
| | - Amit Roy
- Clinical Pharmacology and Pharmacometrics, Research and Development, Bristol-Myers Squibb (United States)
| | - Li Zhu
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb (United States)
| |
Collapse
|
43
|
Optimized Dosing: The Next Step in Precision Medicine in Non-Small-Cell Lung Cancer. Drugs 2021; 82:15-32. [PMID: 34894338 DOI: 10.1007/s40265-021-01654-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
In oncology, and especially in the treatment of non-small-cell lung cancer (NSCLC), dose optimization is often a neglected part of precision medicine. Many drugs are still being administered in "one dose fits all" regimens or based on parameters that are often only minor determinants for systemic exposure. These dosing approaches often introduce additional pharmacokinetic variability and do not add to treatment outcomes. Fortunately, pharmacological knowledge is increasing, providing valuable information regarding the potential of, for example, therapeutic drug monitoring. This article focuses on the evidence for the most promising and easily implemented optimized dosing approaches for the small-molecule inhibitors, chemotherapeutic agents, and monoclonal antibodies as treatment options currently approved for NSCLC. Despite limitations such as investigations having been conducted in oncological diseases other than NSCLC or the retrospective origin of many analyses, an alternative dosing regimen could be beneficial for treatment outcomes, prescriber convenience, or financial burden on healthcare systems. This review of the literature provides recommendations on the implementation of dose optimization and advice regarding promising strategies that deserve further research in NSCLC.
Collapse
|
44
|
Peters S, Galle PR, Bernaards CA, Ballinger M, Bruno R, Quarmby V, Ruppel J, Vilimovskij A, Wu B, Sternheim N, Reck M. Evaluation of atezolizumab immunogenicity: Efficacy and safety (Part 2). Clin Transl Sci 2021; 15:141-157. [PMID: 34582105 PMCID: PMC8742640 DOI: 10.1111/cts.13149] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/07/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023] Open
Abstract
Antibody therapeutics can be associated with unwanted immune responses resulting in the development of anti‐drug antibodies (ADA). Optimal methods to evaluate the potential effects of ADA on clinical outcomes in oncology are not well established. In this study, we assessed efficacy and safety, based on ADA status, in patients from over 10 clinical trials that evaluated the immune checkpoint inhibitor atezolizumab as a single agent or as combination therapy for several types of advanced cancers. ADA can only be observed post randomization, and imbalances in baseline prognostic factors can confound the interpretation of ADA impact. We applied methodology to account for the confounding effects of baseline clinical characteristics and survivorship bias on efficacy. Adjusted meta‐analyses revealed that despite numerical differences in overall survival and progression‐free survival between ADA‐positive and ADA‐negative patients from some studies, ADA‐positive patients from studies with an overall treatment effect derived benefit from atezolizumab, compared with their adjusted controls. Based on large, pooled populations from atezolizumab monotherapy or combination studies, unadjusted descriptive analyses did not identify a clear relationship between ADA status and frequency or severity of adverse events. Data also suggested that any ADA impact is not driven by neutralizing activity. Collectively, this exploratory analysis suggests that the potential for ADA development should not impact treatment decisions with atezolizumab.
Collapse
Affiliation(s)
- Solange Peters
- Oncology Department - CHUV, Lausanne University, Lausanne, Switzerland
| | - Peter R Galle
- Department of Internal Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Coen A Bernaards
- Product Development Biometrics, Genentech, Inc, South San Francisco, California, USA
| | - Marcus Ballinger
- Product Development Oncology, Genentech, Inc, South San Francisco, California, USA
| | - René Bruno
- Clinical Pharmacology, Genentech-Roche, Marseille, France
| | - Valerie Quarmby
- BioAnalytical Sciences, Genentech Inc, South San Francisco, California, USA
| | - Jane Ruppel
- BioAnalytical Sciences, Genentech Inc, South San Francisco, California, USA
| | | | - Benjamin Wu
- Department of Clinical Pharmacology, Genentech Inc, South San Francisco, California, USA
| | - Nitzan Sternheim
- Product Development Regulatory, Genentech Inc, South San Francisco, California, USA
| | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| |
Collapse
|
45
|
Bansal A, Pandey MK, Barham W, Liu X, Harrington SM, Lucien F, Dong H, Park SS, DeGrado TR. Non-invasive immunoPET imaging of PD-L1 using anti-PD-L1-B11 in breast cancer and melanoma tumor model. Nucl Med Biol 2021; 100-101:4-11. [PMID: 34119742 DOI: 10.1016/j.nucmedbio.2021.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Immunotherapy targeting PD-1/PD-L1 immune checkpoint inhibition (ICI) is efficacious in various solid and hematologic malignancies. However, the response rate to PD-1/PD-L1 therapy is only 15-35%. To obtain optimal clinical response to ICI therapies, a reliable assessment of tumor PD-L1 expression is needed to select appropriate patients, and a non-invasive imaging-based assessment of PD-L1 expression is critically needed. Although radiolabeled PET probes based on PD-L1 targeted therapeutic antibodies (e.g. atezolizumab) have shown encouraging results, there is concern that residual therapeutic antibody may compete for binding with the radiotracer thereby compromising imaging studies that follow treatment. METHODS AND RESULTS In this study, we used novel anti-PD-L1-B11 clone antibody known to bind to a different epitope of PD-L1 than the therapeutic antibodies to avoid potential saturation effects. The anti-PD-L1-B11 clone was radiolabeled with zirconium-89 and evaluated to detect PD-L1 expression in various in vitro and in vivo cancer model systems in comparison with [89Zr]Zr-DFO-NCS-atezolizumab. In vitro binding parameters of anti-PD-L1-B11 were like those of atezolizumab. [89Zr]Zr-DFO-NCS-anti-PD-L1-B11 clone showed favorable properties to [89Zr]Zr-DFO-NCS-atezolizumab in an in vivo breast cancer tumor model system with higher uptake in PD-L1 expressing tumors. CONCLUSION Our data demonstrates that [89Zr]Zr-DFO-NCS-anti-PD-L1-B11 exhibits excellent imaging properties for the assessment PD-L1 expression. The independent binding site of anti-PD-L1-B11 relative to therapeutic anti-PD-L1 antibodies may be advantageous for anti-PD-L1 therapy monitoring.
Collapse
Affiliation(s)
- Aditya Bansal
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| | | | - Whitney Barham
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Xin Liu
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN, USA; Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
46
|
Wu B, Sternheim N, Agarwal P, Suchomel J, Vadhavkar S, Bruno R, Ballinger M, Bernaards CA, Chan P, Ruppel J, Jin J, Girish S, Joshi A, Quarmby V. Evaluation of atezolizumab immunogenicity: Clinical pharmacology (part 1). Clin Transl Sci 2021; 15:130-140. [PMID: 34432389 PMCID: PMC8742635 DOI: 10.1111/cts.13127] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/07/2021] [Accepted: 07/19/2021] [Indexed: 01/10/2023] Open
Abstract
Baseline patient characteristics and prognostic factors are important considerations in oncology when evaluating the impact of immunogenicity on pharmacokinetics (PK) and efficacy. Here, we assessed the impact of anti-drug antibodies (ADA) on the PK of the immune checkpoint inhibitor atezolizumab (an anti-PD-L1 monoclonal antibody). We evaluated data from ≈ 4500 patients from 12 clinical trials across different tumor types, treatment settings, and dosing regimens. In our dataset, ~ 30% of patients (range, 13-54%) developed treatment-emergent ADA, and in vitro neutralizing antibodies (NAb) were seen in ~ 50% of ADA-positive (+) patients. Pooled time course data showed a trend toward lower atezolizumab exposure in ADA+ patients, which was more pronounced in ADA+/NAb+ patients. However, the atezolizumab concentration distributions overlapped, and drug concentrations exceeded 6 µg/ml, the target concentration required for receptor saturation, in greater than 95% of patients. Patients had sufficient exposure regardless of ADA status. The dose selected to allow for dosing over effects from ADA resulted in a flat exposure-response relationship. Analysis of study results by ADA titer showed that exposure and overall survival were not affected in a clinically meaningful way. High tumor burden, low albumin, and high CRP at baseline showed the greatest association with ADA development but not with subsequent NAb development. These imbalanced factors at baseline can confound analysis of ADA impact. ADA increases atezolizumab clearance minimally (9%), and its impact on exposure based on the totality of the clinical pharmacology assessment does not appear to be clinically meaningful.
Collapse
Affiliation(s)
- Benjamin Wu
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Nitzan Sternheim
- Product Development Regulatory, Genentech Inc., South San Francisco, California, USA
| | - Priya Agarwal
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Julia Suchomel
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Shweta Vadhavkar
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Rene Bruno
- Clinical Pharmacology, Genentech-Roche, Marseille, France
| | - Marcus Ballinger
- Product Development Oncology Clinical Sciences, Genentech Inc., South San Francisco, California, USA
| | - Coen A Bernaards
- Product Development, Biostatistics, Genentech Inc., South San Francisco, California, USA
| | - Phyllis Chan
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Jane Ruppel
- BioAnalytical Sciences, Genentech Inc., South San Francisco, California, USA
| | - Jin Jin
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Sandhya Girish
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Amita Joshi
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Valerie Quarmby
- BioAnalytical Sciences, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
47
|
Shemesh CS, Chan P, Shao H, Xu DZ, Combs D, Vadhavkar S, Bruno R, Wu B. Atezolizumab and Bevacizumab in Patients with Unresectable Hepatocellular Carcinoma: Pharmacokinetic and Safety Assessments Based on Hepatic Impairment Status and Geographic Region. Liver Cancer 2021; 10:485-499. [PMID: 34721510 PMCID: PMC8527900 DOI: 10.1159/000515817] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/11/2021] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Phase 1b GO30140 and phase 3 IMbrave150 studies evaluated first-line atezolizumab + bevacizumab for unresectable hepatocellular carcinoma (HCC). Here, we evaluated pharmacokinetics (PK) and safety by hepatic impairment status and geographic region. METHODS Patients received atezolizumab 1,200 mg + bevacizumab 15 mg/kg IV every 3 weeks. Drug concentrations were evaluated by descriptive statistics and population PK. PK and adverse event frequencies were evaluated by hepatic impairment status and region. RESULTS 323 IMbrave150 patients and 162 GO30140 patients were PK evaluable. Compared with IMbrave150 patients who had normal hepatic function per the National Cancer Institute Organ Dysfunction Working Group (NCI-ODWG) criteria (n = 123), patients with mild impairment (n = 171) had a geometric mean ratio (GMR) of 0.92 for cycle 1 atezolizumab area under the concentration-time curve (AUC); patients with moderate impairment (n = 27) had a GMR of 0.88. Patients in Asia ([n = 162] vs. outside [n = 161]) had a GMR of 1.25 for cycle 1 atezolizumab AUC. Compared with GO30140 patients who had normal hepatic function (NCI-ODWG [n = 61]), patients with mild impairment (n = 92) had a GMR of 0.97 for cycle 1 peak bevacizumab concentrations; those with moderate impairment (n = 9) had a GMR of 0.94. Patients in Asia (n = 111) versus outside Asia (n = 51) had a GMR of 0.94 for cycle 1 peak bevacizumab concentration. PK results were generally comparable when evaluated based on additional hepatic functional definitions (Child-Pugh or albumin/bilirubin criteria) or study enrollment in Japan. No associations between atezolizumab PK and HCC etiology were seen. Adverse event frequencies were similar across evaluated groups. CONCLUSIONS IMbrave150 and GO30140 patients with unresectable HCC had varying baseline hepatic impairment and high enrollment from Asia. PK data demonstrated considerable exposure overlap across groups. Treatment was tolerable across groups. No need for dose adjustment based on mild or moderate hepatic impairment or region is recommended based on this analysis.
Collapse
Affiliation(s)
- Colby S. Shemesh
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA,*Colby S. Shemesh,
| | - Phyllis Chan
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Hui Shao
- Safety Science, F. Hoffmann-La Roche Ltd., Beijing, China
| | - Derek-Zhen Xu
- Product Development Oncology, F. Hoffmann-La Roche Ltd., Shanghai, China
| | - Daniel Combs
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - Shweta Vadhavkar
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| | - René Bruno
- Clinical Pharmacology, Genentech-Roche, Marseille, France
| | - Benjamin Wu
- Department of Clinical Pharmacology, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
48
|
Evaluating the efficacy and safety of immune checkpoint inhibitors by detecting the exposure-response: An inductive review. Int Immunopharmacol 2021; 97:107703. [PMID: 33933843 DOI: 10.1016/j.intimp.2021.107703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have been demonstrated an effective treatment in multiple tumor type, which restore the immune response to against cancer cell. Currently, approved ICIs include anti-cytotoxic T-lymphocyte antigen-4 (CTLA-4); anti-programmed cell death 1 (PD-1) and anti-programmed cell death ligand 1 (PD-L1) monoclonal antibodies (mAbs). In most these drugs, unique pharmacokinetic (PK) and pharmacodynamics (PD) have shown significant influence on clinical outcomes, which occurred by target-mediated drug concentration and time-varying drug clearance. An exposure-response (E-R) relationship has been used to describe the safety and efficacy of ICIs, and shown a plateaued E-R and time dependent changes in exposure. Using an enzyme linked immunosorbent assay (ELISA) or LC-MS/MS method to measure the peak concentration, trough concentration or area under the curve (AUC) of ICIs to assess the drug exposure. There are lots of covariates that have an influence on exposure, such as sex, clearance, body weight and tumor burden. In this review, we pooled data from studies of concentration or other pharmacokinetics parameter of mAbs to assess E-R in efficacy and safety.
Collapse
|
49
|
Marchand M, Zhang R, Chan P, Quarmby V, Ballinger M, Sternheim N, Wu B, Jin JY, Bruno R. Time-dependent population PK models of single-agent atezolizumab in patients with cancer. Cancer Chemother Pharmacol 2021; 88:211-221. [PMID: 33904970 DOI: 10.1007/s00280-021-04276-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/09/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE The time-varying clearance (CL) of the PD-L1 inhibitor atezolizumab was assessed on a population of 1519 cancer patients (primarily with non-small-cell lung cancer or metastatic urothelial carcinoma) from three clinical studies. METHODS The first step was to identify the baseline covariates affecting atezolizumab CL without including time-varying components (stationary covariate model). Two time-varying models were then investigated: (1) a model allowing baseline covariates to vary over time (time-varying covariate model), (2) a model with empirical time-varying Emax CL function. RESULTS The final stationary covariate model included main effects of body weight, albumin levels, tumor size, anti-drug antibodies (ADA) and gender on atezolizumab CL. Both time-varying models resulted in a clear improvement of the data fit and visual predictive checks over the stationary model. The time-varying covariate model provided the best fit of the data. In this model, the main driver for change in CL over time was variations in albumin level with an increase in serum albumin (improvement in a patient's status) mirroring a decrease in CL. Time-varying ADAs had a small impact (9% increase in CL). None of the covariates impacted atezolizumab CL by more than ± 30% from median. The estimated maximum decrease in CL with time was 22% with the Emax model. CONCLUSION The overall impact of covariates on atezolizumab CL did not warrant any change in atezolizumab dosing recommendations. The results support the hypothesis that variation in atezolizumab CL over time is associated with patients' disease status, as shown with other checkpoint inhibitors.
Collapse
Affiliation(s)
- Mathilde Marchand
- Certara Strategic Consulting, Certara, 54 Rue de Londres, 75009, Paris, France
| | - Rong Zhang
- Clinical Pharmacology, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Phyllis Chan
- Clinical Pharmacology, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Valerie Quarmby
- BioAnalytical Sciences, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Marcus Ballinger
- Product Development, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Nitzan Sternheim
- Product Development, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jin Y Jin
- Clinical Pharmacology, Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - René Bruno
- Clinical Pharmacology, Genentech-Roche, 84 Chemin des Grives, 13013, Marseille, France.
| |
Collapse
|
50
|
Chatelut E, Hendrikx JJMA, Martin J, Ciccolini J, Moes DJAR. Unraveling the complexity of therapeutic drug monitoring for monoclonal antibody therapies to individualize dose in oncology. Pharmacol Res Perspect 2021; 9:e00757. [PMID: 33745217 PMCID: PMC7981594 DOI: 10.1002/prp2.757] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Monoclonal antibodies (Mabs) have become key drugs in cancer treatment, either as targeted therapies or more recently as immune checkpoint inhibitors (ICIs). The fact that only some patients benefit from these drugs poses the usual question in the field of onco-hematology: that of the benefit of individual dosing and the potential of therapeutic drug monitoring (TDM) to carry out this individualization. However, Mabs present unique pharmacological characteristics for TDM, and the pharmacokinetic-pharmacodynamic relationship observed should be interpreted differently than that observed for conventional drugs and small molecules. This pharmacology practice review has been summarized from a public debate between the authors at the International TDM and Clinical Toxicology meeting in Banff, 2020, regarding the potential roles of TDM in the Mab/ICI setting.
Collapse
Affiliation(s)
- Etienne Chatelut
- CRCTUniversité de ToulouseInserm, and Institut Claudius‐RegaudIUCT‐OncopoleToulouseFrance
| | - Jeroen J. M. A. Hendrikx
- Department of Pharmacy and Pharmacology and Department of Nuclear MedicineThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jennifer Martin
- Centre for Drug Repurposing and Medicines ResearchThe University of NewcastleCallaghanNSWAustralia
| | - Joseph Ciccolini
- SMARTcCRCM Inserm U1068Aix Marseille University and La Timone university Hospital of MarseilleMarseilleFrance
| | - Dirk Jan A. R. Moes
- Department of Clinical Pharmacy and ToxicologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|