1
|
Peribañez-Dominguez S, Parra-Guillen Z, Troconiz IF. Development and Application of Physiologically-Based Pharmacokinetic Model to Predict Systemic and Organ Exposure of Colorectal Cancer Drugs. Pharmaceutics 2025; 17:57. [PMID: 39861705 PMCID: PMC11768185 DOI: 10.3390/pharmaceutics17010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/23/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Colorectal cancer (CRC) holds the third and second position among cancers affecting men and women, respectively. Frequently, the first-line treatment for metastatic CRC consists of the intravenous administration of 5-fluorouracil and leucovorin in combination with oxaliplatin or irinotecan. Physiologically-based pharmacokinetic models (PBPK) aim to mechanistically incorporate body physiology and drug physicochemical attributes, enabling the description of both systemic and organ drug exposure based on the treatment specificities. This bottom-up approach represents an opportunity to personalize treatment and minimize the therapeutic risk/benefit ratio through the understanding of drug distribution within colorectal tissue. This project has the goal of characterizing the systemic and tissue exposure of four anti-cancer drugs in humans using a PBPK platform fed with data from the literature. METHODS A literature search was performed to collect clinical data on systemic concentration versus time profiles. Physicochemical features were obtained from the literature, as well as parameters associated with distribution, metabolism, and excretion. The PBPK models were built using PK-Sim®. RESULTS The data from 51 clinical studies were extracted and combined in one single dataset. The PBPK models successfully described the exposure vs. time profiles with respect to both, with both the typical tendency and dispersion shown by the data. The percentage of observations falling within the two-fold error bounds ranged between 94 and 100%. The colon/plasma AUCinf ratios were similar for 5-FU, oxaliplatin, and leucovorin, but it was significantly higher for irinotecan. CONCLUSIONS The PBPK models support tailored treatment approaches by linking in vitro studies to organ exposure. These models serve as the initial step towards incorporating a dedicated tumor compartment, which will further account for the variability in tumor microenvironment characteristics to improve therapeutic strategies.
Collapse
Affiliation(s)
- Sara Peribañez-Dominguez
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, 31009 Pamplona, Spain; (Z.P.-G.); (I.F.T.)
- Navarra Institute for Health Research (IdiSNA), 31002 Pamplona, Spain
| | - Zinnia Parra-Guillen
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, 31009 Pamplona, Spain; (Z.P.-G.); (I.F.T.)
- Navarra Institute for Health Research (IdiSNA), 31002 Pamplona, Spain
| | - Iñaki F. Troconiz
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, 31009 Pamplona, Spain; (Z.P.-G.); (I.F.T.)
- Navarra Institute for Health Research (IdiSNA), 31002 Pamplona, Spain
- Navarra Institute of Data Science and Artificial Intelligence, DATAI, University of Navarra, 31009 Pamplona, Spain
| |
Collapse
|
2
|
Kim MK, Cho IR, Kim Y, Choi JH, Jung K, Kim J, Kim S, Yun H, Yoon J, Oh DY, Kim K, Lee SH. Prognostic value of the TP53 mutation in patients with pancreatic ductal adenocarcinoma receiving FOLFIRINOX. Ther Adv Med Oncol 2024; 16:17588359241290482. [PMID: 39449732 PMCID: PMC11500227 DOI: 10.1177/17588359241290482] [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: 04/25/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Background KRAS, TP53, CDKN2A, and SMAD4 have been the main driver mutations in pancreatic ductal adenocarcinoma (PDAC). Studies on the clinical significance and treatment response to 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) regimen in terms of the presence of these mutations remain inconclusive. Objectives This study aimed to compare the survival outcome and response to FOLFIRINOX chemotherapy based on the presence of four driver mutation genes. Design A multi-center retrospective study conducted at two tertiary medical centers. Methods This study analyzed PDAC patients who were treated with FOLFIRINOX chemotherapy as the initial treatment. Tumor specimens were analyzed by a targeted next-generation sequencing platform at two tertiary referral hospitals from January 2016 to March 2022. Patients' demographics, survival outcomes, and chemotherapeutic response were investigated and compared according to the presence of driver mutations. Results The analysis included 100 patients. KRAS mutation was identified in 92 (92.0%) patients, followed by TP53, CDKN2A, and SMAD4 in 63 (63.0%), 18 (18.0%), and 17 (17.0%) patients, respectively. The TP53 wild-type group demonstrated longer overall survival (OS) than the TP53 mutated group (median OS: 29 vs 19 months, p = 0.03), and TP53 served as a prognostic factor for survival (hazard ratio = 1.74, 95% confidence interval: 1.00-3.00, p = 0.048). The difference in OS according to TP53 mutation was intensified in localized pancreatic adenocarcinoma (37 vs 19 months, p = 0.01). The TP53 wild-type group demonstrated a higher objective response rate to FOLFIRINOX chemotherapy than the TP53 mutation group in localized pancreatic adenocarcinoma (50.0% vs 17.6%, p = 0.024). Conclusion PDAC patients with wild-type TP53 demonstrated longer OS than those with TP53 mutation, and this trend was intensified in patients with localized disease. This result may be due to an impaired response to FOLFIRINOX chemotherapy in patients with TP53 mutation.
Collapse
Affiliation(s)
- Min Kyu Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In Rae Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yooeun Kim
- Interdisciplinary Program in Bioinformatics, College of Natural Science, Seoul National University, Seoul, Republic of Korea
| | - Jin Ho Choi
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Sheehyun Kim
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hongseok Yun
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeesun Yoon
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwangsoo Kim
- Department of Transdisciplinary Medicine, Institute of Convergence Medicine with Innovative Technology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
3
|
Willems RAL, Michiels N, Lanting VR, Bouwense S, van den Broek BLJ, Graus M, Klok FA, Groot Koerkamp B, de Laat B, Roest M, Wilmink JW, van Es N, Mieog JSD, Ten Cate H, de Vos-Geelen J. Venous Thromboembolism and Primary Thromboprophylaxis in Perioperative Pancreatic Cancer Care. Cancers (Basel) 2023; 15:3546. [PMID: 37509209 PMCID: PMC10376958 DOI: 10.3390/cancers15143546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Recent studies have shown that patients with pancreatic ductal adenocarcinoma (PDAC) treated with neoadjuvant chemo(radio)therapy followed by surgery have an improved outcome compared to patients treated with upfront surgery. Hence, patients with PDAC are more and more frequently treated with chemotherapy in the neoadjuvant setting. PDAC patients are at a high risk of developing venous thromboembolism (VTE), which is associated with decreased survival rates. As patients with PDAC were historically offered immediate surgical resection, data on VTE incidence and associated preoperative risk factors are scarce. Current guidelines recommend primary prophylactic anticoagulation in selected groups of patients with advanced PDAC. However, recommendations for patients with (borderline) resectable PDAC treated with chemotherapy in the neoadjuvant setting are lacking. Nevertheless, the prevention of complications is crucial to maintain the best possible condition for surgery. This narrative review summarizes current literature on VTE incidence, associated risk factors, risk assessment tools, and primary thromboprophylaxis in PDAC patients treated with neoadjuvant chemo(radio)therapy.
Collapse
Affiliation(s)
- R A L Willems
- Department of Functional Coagulation, Synapse Research Institute, 6217 KD Maastricht, The Netherlands
- Thrombosis Expert Center Maastricht, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- Department of Internal Medicine, Section Vascular Medicine, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- Department of Internal Medicine, Section Medical Oncology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- CARIM, School for Cardiovascular Diseases, 6229 ER Maastricht, The Netherlands
| | - N Michiels
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - V R Lanting
- Department of Internal Medicine, Section Vascular Medicine, University of Amsterdam, Amsterdam UMC Location, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, 1081 HV Amsterdam, The Netherlands
- Tergooi Hospitals, Internal Medicine, 1201 DA Hilversum, The Netherlands
| | - S Bouwense
- Department of Surgery, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- NUTRIM, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| | - B L J van den Broek
- Department of Surgery, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - M Graus
- Department of Internal Medicine, Section Medical Oncology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- GROW, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| | - F A Klok
- Department of Medicine-Thrombosis and Hemostasis, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - B Groot Koerkamp
- Department of Surgery, Erasmus University Medical Center, 3015 CN Rotterdam, The Netherlands
| | - B de Laat
- Department of Functional Coagulation, Synapse Research Institute, 6217 KD Maastricht, The Netherlands
- CARIM, School for Cardiovascular Diseases, 6229 ER Maastricht, The Netherlands
- Department of Platelet Pathophysiology, Synapse Research Institute, 6217 KD Maastricht, The Netherlands
| | - M Roest
- Department of Platelet Pathophysiology, Synapse Research Institute, 6217 KD Maastricht, The Netherlands
| | - J W Wilmink
- Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam University Medical Center, Location University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - N van Es
- Department of Internal Medicine, Section Vascular Medicine, University of Amsterdam, Amsterdam UMC Location, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, 1081 HV Amsterdam, The Netherlands
| | - J S D Mieog
- Department of Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - H Ten Cate
- Thrombosis Expert Center Maastricht, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- Department of Internal Medicine, Section Vascular Medicine, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- CARIM, School for Cardiovascular Diseases, 6229 ER Maastricht, The Netherlands
| | - J de Vos-Geelen
- Department of Internal Medicine, Section Medical Oncology, Maastricht University Medical Center, 6202 AZ Maastricht, The Netherlands
- GROW, Maastricht University Medical Center, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
4
|
Sex and Circadian Timing Modulate Oxaliplatin Hematological and Hematopoietic Toxicities. Pharmaceutics 2022; 14:pharmaceutics14112465. [PMID: 36432655 PMCID: PMC9699532 DOI: 10.3390/pharmaceutics14112465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 11/17/2022] Open
Abstract
Oxaliplatin was nearly twice as hematotoxic, with optimal circadian timing differing by 6 h, in women as compared to men with colorectal cancers. Hence, we investigated sex- and timing-related determinants of oxaliplatin hematopoietic toxicities in mice. Body-weight loss (BWL), blood cell counts, bone marrow cellularity (BMC) and seven flow-cytometry-monitored hematopoietic progenitor populations were evaluated 72 h after oxaliplatin chronotherapy administration (5 mg/kg). In control animals, circadian rhythms of circulating white blood cells showed a peak at ZT5 in both sexes, whereas BMC was maximum at ZT20 in males and ZT13h40 in females. All BM progenitor counts presented robust rhythms with phases around ZT3h30 in females, whereas only three of them rhythmically cycled in males with a ≈ -6 h phase shift. In treated females, chronotoxicity rhythms occurred in BWL, WBC, BMC and all BM progenitors with the best timing at ZT15, ZT21, ZT15h15 and ZT14h45, respectively. In males, almost no endpoints showed circadian rhythms, BWL and WBC toxicity being minimal, albeit with a substantial drop in BM progenitors. Increasing dose (10 mg/kg) in males induced circadian rhythms in BWL and WBC but not in BM endpoints. Our results suggest complex and sex-specific clock-controlled regulation of the hematopoietic system and its response to oxaliplatin.
Collapse
|
5
|
Sally Á, McGowan R, Finn K, Moran BM. Current and Future Therapies for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14102417. [PMID: 35626020 PMCID: PMC9139531 DOI: 10.3390/cancers14102417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/28/2022] [Accepted: 05/11/2022] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Pancreatic cancer is the fourth leading cause of cancer-related mortality worldwide. The poor survival associated with this disease is due to delayed diagnosis, a lack of reliable biomarkers, and tumour resistance to treatment. Currently, surgery is the only curative treatment option, but few patients are eligible for this procedure. Developing resistance to current chemotherapies such as gemcitabine has led to a reduction in effective therapy options for patients and an urgent requirement for the development of novel therapeutic avenues. Potential success has been noted in therapeutic approaches such as synthetic lethality and immunotherapy. An array of clinical trials are currently recruiting, primarily in the area of monoclonal antibodies in combination with other therapies such as chemotherapy and immune checkpoint inhibitors. This review article aims to highlight the potential these therapies have to improve patient prognosis and survival. Abstract Pancreatic cancer is one of the leading causes of cancer-related death worldwide. This is due to delayed diagnosis and resistance to traditional chemotherapy. Delayed diagnosis is often due to the broad range of non-specific symptoms that are associated with the disease. Resistance to current chemotherapies, such as gemcitabine, develops due to genetic mutations that are either intrinsic or acquired. This has resulted in poor patient prognosis and, therefore, justifies the requirement for new targeted therapies. A synthetic lethality approach, that targets specific loss-of-function mutations in cancer cells, has shown great potential in pancreatic ductal adenocarcinoma (PDAC). Immunotherapies have also yielded promising results in the development of new treatment options, with several currently undergoing clinical trials. The utilisation of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell transfer, and vaccines have shown success in several neoplasms such as breast cancer and B-cell malignancies and, therefore, could hold the same potential in PDAC treatment. These therapeutic strategies could have the potential to be at the forefront of pancreatic cancer therapy in the future. This review focuses on currently approved therapies for PDAC, the challenges associated with them, and future directions of therapy including synthetically lethal approaches, immunotherapy, and current clinical trials.
Collapse
Affiliation(s)
- Áine Sally
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
| | - Ryan McGowan
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
- Department of Life Sciences, School of Science, Atlantic Technological University Sligo, Ash Lane, Ballytivnan, F91 YW50 Sligo, Ireland
| | - Karen Finn
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
| | - Brian Michael Moran
- Department of Analytical, Biopharmaceutical and Medical Sciences, School of Science and Computing, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland; (Á.S.); (R.M.); (K.F.)
- Correspondence:
| |
Collapse
|
6
|
Brendel K, Bekaii‐Saab T, Boland PM, Dayyani F, Dean A, Macarulla T, Maxwell F, Mody K, Pedret‐Dunn A, Wainberg ZA, Zhang B. Population pharmacokinetics of liposomal irinotecan in patients with cancer and exposure–safety analyses in patients with metastatic pancreatic cancer. CPT Pharmacometrics Syst Pharmacol 2021; 10:1550-1563. [PMID: 34750990 PMCID: PMC8674005 DOI: 10.1002/psp4.12725] [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: 01/15/2021] [Revised: 06/23/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022] Open
Abstract
Liposomal irinotecan is a liposomal formulation of irinotecan, which prolongs circulation of irinotecan and its active metabolite SN‐38. A population pharmacokinetic (PK) model was developed based on data from seven studies (N = 440). Adequacy of the model was assessed using multiple methods, including visual predictive check. Associations between PK exposure and the incidence of diarrhea (grade ≥3) and neutropenia adverse events (AEs) (grade ≥3) at first event in patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) were investigated using logistic regression based on data from two studies (the phase III NAPOLI‐1 [N = 260] and phase I/II NCT02551991 [N = 56] trials). The PKs of total irinotecan was described by a two‐compartment model with first‐order elimination, with SN‐38 formed directly by a first‐order constant from the central compartment of irinotecan or after using a transit compartment. Clearance was 17.9 L/week (0.107 L/h) and 19,800 L/week (118 L/h) for total irinotecan and SN‐38, respectively. The UGT1A1*28 7/7 homozygous genotype had no significant impact on SN‐38 clearance. Model evaluation was satisfactory for both irinotecan and SN‐38. The incidence of diarrhea (grade ≥3) at first event was significantly higher with increasing average concentrations of total irinotecan and SN‐38; there was no significant association between an increased risk of neutropenia AEs (grade ≥3) at first event and average SN‐38 concentrations. In summary, the PKs of total irinotecan and SN‐38 after administration of liposomal irinotecan were well‐described by the model. The UGT1A1*28 status had no significant impact on the PKs of liposomal irinotecan.
Collapse
Affiliation(s)
| | | | | | | | - Andrew Dean
- St John of God Hospital Subiaco Perth Western Australia Australia
| | - Teresa Macarulla
- Vall d´Hebrón University Hospital Vall d´Hebrón Institute of Oncology Barcelona Spain
| | | | | | | | - Zev A Wainberg
- Ronald Regan UCLA Medical Center Los Angeles California USA
| | | |
Collapse
|
7
|
Wang H, Ding W, Shi H, Bao H, Lu Y, Jiang TA. Combination therapy with low-frequency ultrasound irradiation and radiofrequency ablation as a synergistic treatment for pancreatic cancer. Bioengineered 2021; 12:9832-9846. [PMID: 34696663 PMCID: PMC8810087 DOI: 10.1080/21655979.2021.1995581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We aim to evaluate the efficacies of combination therapy with low-frequency ultrasound-stimulated microbubbles (USMB) and radiofrequency ablation (RFA) on suppressing the proliferation of pancreatic cancer cell and treating Panc02 subcutaneous xenograft mice. The proliferation of HPDE6-C7 and Panc02 cells after the treatment of USMB and RFA alone or combination were evaluated by CCK-8 assay. Scratch test was performed to assess the cell migration capability. Panc02-bearing mice were received 14-day treatment of USMB and RFA alone or combination. Tumor size and survival rate were recorded once two days. The serum levels of immune-related factors and changes of apoptosis- and autophagy-related factors were detected by ELISA and western blotting methods. As a result, CKK-8 assays revealed significant inhibition on Panc02 cell proliferation in combination therapy with USMB and RFA relative to other groups (all p < 0.05). Strong synergistic effect of USMB combined with RFA was confirmed via the calculated combination index (CI) <0.4. In addition, combination therapy of USMB and RFA significantly inhibited the migration of Panc02 cells. Moreover, combined treatment remarkably inhibited the size and width of xenograft and improved the survival in Panc02-bearing mice. Furthermore, 14-day combination therapy of USMB and RFA in Panc02-bearing mice significantly facilitated the apoptosis and autophagy of tumor cells. In summary, combination therapy of USMB and RFA showed synergistic anti-tumor efficacies on Panc02 cells attributing to the promotion on apoptosis and autophagy in Panc02 subcutaneous xenograft mice.
Collapse
Affiliation(s)
- Huiyang Wang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenxiu Ding
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongwei Shi
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiwei Bao
- Department of Ultrasound Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuting Lu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian An Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pulsed Electric Field Technology for Medical Transformation, Hangzhou, China
| |
Collapse
|
8
|
Hadj Bachir E, Poiraud C, Paget S, Stoup N, El Moghrabi S, Duchêne B, Jouy N, Bongiovanni A, Tardivel M, Weiswald LB, Vandepeutte M, Beugniez C, Escande F, Leteurtre E, Poulain L, Lagadec C, Pigny P, Jonckheere N, Renaud F, Truant S, Van Seuningen I, Vincent A. A new pancreatic adenocarcinoma-derived organoid model of acquired chemoresistance to FOLFIRINOX: First insight of the underlying mechanisms. Biol Cell 2021; 114:32-55. [PMID: 34561874 DOI: 10.1111/boc.202100003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND INFORMATION Although improvements have been made in the management of pancreatic adenocarcinoma (PDAC) during the past 20 years, the prognosis of this deadly disease remains poor with an overall 5-year survival under 10%. Treatment with FOLFIRINOX, a combined regimen of 5-fluorouracil, irinotecan (SN-38) and oxaliplatin, is nonetheless associated with an excellent initial tumour response and its use has allowed numerous patients to go through surgery while their tumour was initially considered unresectable. These discrepancies between initial tumour response and very low long-term survival are the consequences of rapidly acquired chemoresistance and represent a major therapeutic frontier. To our knowledge, a model of resistance to the combined three drugs has never been described due to the difficulty of modelling the FOLFIRINOX protocol both in vitro and in vivo. Patient-derived tumour organoids (PDO) are the missing link that has long been lacking in the wide range of epithelial cancer models between 2D adherent cultures and in vivo xenografts. In this work we sought to set up a model of PDO with resistance to FOLFIRINOX regimen that we could compare to the paired naive PDO. RESULTS We first extrapolated physiological concentrations of the three drugs using previous pharmacodynamics studies and bi-compartmental elimination models of oxaliplatin and SN-38. We then treated PaTa-1818x naive PDAC organoids with six cycles of 72 h-FOLFIRINOX treatment followed by 96 h interruption. Thereafter, we systematically compared treated organoids to PaTa-1818x naive organoids in terms of growth, proliferation, viability and expression of genes involved in cancer stemness and aggressiveness. CONCLUSIONS We reproductively obtained resistant organoids FoxR that significantly showed less sensitivity to FOLFORINOX treatment than the PaTa-1818x naive organoids from which they were derived. Our resistant model is representative of the sequential steps of chemoresistance observed in patients in terms of growth arrest (proliferation blockade), residual disease (cell quiescence/dormancy) and relapse. SIGNIFICANCE To our knowledge, this is the first genuine in vitro model of resistance to the three drugs in combined therapy. This new PDO model will be a great asset for the discovery of acquired chemoresistance mechanisms, knowledge that is mandatory before offering new therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Elsa Hadj Bachir
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Charles Poiraud
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Digestive Surgery and Transplantation, CHU Lille, Lille, France
| | - Sonia Paget
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Nicolas Stoup
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Soumaya El Moghrabi
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Belinda Duchêne
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Nathalie Jouy
- UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, BioImaging Center Lille (BICeL), Univ. Lille, Lille, France
| | - Antonino Bongiovanni
- UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, BioImaging Center Lille (BICeL), Univ. Lille, Lille, France
| | - Meryem Tardivel
- UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, BioImaging Center Lille (BICeL), Univ. Lille, Lille, France
| | - Louis-Bastien Weiswald
- UNICAEN, Inserm U1086 ANTICIPE "Interdisciplinary Research Unit for Cancer Prevention and Treatment", Normandie Univ, Caen, France.,Cancer Centre F. Baclesse, UNICANCER, Caen, France
| | - Marie Vandepeutte
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - César Beugniez
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Digestive Surgery and Transplantation, CHU Lille, Lille, France
| | - Fabienne Escande
- Department of Biochemistry and Molecular Biology, CHU Lille, Hormonology Metabolism Nutrition Oncology, Lille, France
| | - Emmanuelle Leteurtre
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Pathology, CHU Lille, Univ. Lille, Lille, France
| | -
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Laurent Poulain
- UNICAEN, Inserm U1086 ANTICIPE "Interdisciplinary Research Unit for Cancer Prevention and Treatment", Normandie Univ, Caen, France.,Cancer Centre F. Baclesse, UNICANCER, Caen, France
| | - Chann Lagadec
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Pascal Pigny
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Nicolas Jonckheere
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Florence Renaud
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Pathology, CHU Lille, Univ. Lille, Lille, France
| | - Stephanie Truant
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Digestive Surgery and Transplantation, CHU Lille, Lille, France
| | - Isabelle Van Seuningen
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Audrey Vincent
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| |
Collapse
|
9
|
Mathonnet M, Vanderstraete M, Bounaix Morand du Puch C, Giraud S, Lautrette C, Ouaissi M, Tabchouri N, Taïbi A, Martin R, Herafa I, Tchalla A, Christou N. ONCOGRAM: study protocol for the evaluation of therapeutic response and survival of metastatic colorectal cancer patients treated according to the guidelines of a chemosensitivity assay, the Oncogramme®. Trials 2021; 22:556. [PMID: 34419125 PMCID: PMC8379769 DOI: 10.1186/s13063-021-05531-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 08/09/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Colorectal cancer is a major public concern, being the second deadliest cancer in the world. Whereas survival is high for localized forms, metastatic colorectal cancer has showed poor prognosis, with a 5-year survival barely surpassing 11%. Conventional chemotherapies against this disease proved their efficiency and remain essential in first-line treatment. However, the large number of authorized protocols complexifies treatment decision. In common practice, such decision is made on an empirical basis, by assessing benefits and risks for the patient. In other words, there is currently no efficient means of predicting the efficacy of any chemotherapy protocol for metastatic colorectal cancer. METHODS/DESIGN The use of a chemosensitivity assay, the Oncogramme®, should help clinicians administer the best chemotherapy regimen to their patients. We hypothesize it would ultimately improve their survival. In this multicentred, prospective trial (ONCOGRAM), eligible patients with metastatic colorectal cancer are randomized to determine whether they will receive an Oncogramme®. For clinicians whose patients benefited from the assay (arm A), results are used as a decision support tool. Patients not undergoing the Oncogramme® procedure are treated according to current practice, without the assistance of the assay (arm B). Primary outcome is 1-year progression-free survival. Secondary outcomes include response rates, as well as 6-month and 1-year survival rates. DISCUSSION This study aims at investigating the clinical utility of the Oncogramme® as a decision support tool for the treatment of patients with metastatic colorectal cancer. If the Oncogramme® positively influenced patient overall survival and/or progression-free survival, it would be of great value for clinicians to implement this assay within the current landscape of personalized medicine tools, which include genomics and biomarker assays. TRIAL REGISTRATION ClinicalTrials.gov identifier NCT03133273 . Registered on April 28, 2017.
Collapse
Affiliation(s)
- Muriel Mathonnet
- Department of Digestive, General and Endocrinology Surgery, Dupuytren University Hospital, 2 Avenue Martin Luther King, Limoges, France
- EA3842 laboratory (CAPTuR: “Contrôle de l’Activation cellulaire, Progression Tumorale et Résistances thérapeutiques”), Limoges Medical School, 2 rue du docteur Marcland, Limoges, France
| | | | | | | | | | - Mehdi Ouaissi
- Department of Digestive, Oncological, Endocrine, Hepato-Biliary, Pancreatic and Liver Transplant Surgery, Trousseau University Hospital, Avenue de la République, Chambray-lès-Tours, France
| | - Nicolas Tabchouri
- Department of Digestive, Oncological, Endocrine, Hepato-Biliary, Pancreatic and Liver Transplant Surgery, Trousseau University Hospital, Avenue de la République, Chambray-lès-Tours, France
| | - Abdelkader Taïbi
- Department of Digestive, General and Endocrinology Surgery, Dupuytren University Hospital, 2 Avenue Martin Luther King, Limoges, France
| | - Renaud Martin
- Research and Innovation Bureau, Dupuytren University Hospital, 2 Avenue Martin Luther King, Limoges, France
| | - Isabelle Herafa
- Centre of Clinical Investigation 1435, Dupuytren University Hospital, 2 Avenue Martin Luther King, Limoges, France
| | - Achille Tchalla
- Department of Clinical Geriatrics, Dupuytren University Hospital, Limoges, France
| | - Niki Christou
- Department of Digestive, General and Endocrinology Surgery, Dupuytren University Hospital, 2 Avenue Martin Luther King, Limoges, France
- EA3842 laboratory (CAPTuR: “Contrôle de l’Activation cellulaire, Progression Tumorale et Résistances thérapeutiques”), Limoges Medical School, 2 rue du docteur Marcland, Limoges, France
| |
Collapse
|
10
|
Vary A, Lebellec L, Di Fiore F, Penel N, Cheymol C, Rad E, El Hajbi F, Lièvre A, Edeline J, Bimbai AM, Le Deley MC, Turpin A. FOLFIRINOX relative dose intensity and disease control in advanced pancreatic adenocarcinoma. Ther Adv Med Oncol 2021; 13:17588359211029825. [PMID: 34349842 PMCID: PMC8287268 DOI: 10.1177/17588359211029825] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/14/2021] [Indexed: 01/05/2023] Open
Abstract
Background: Most patients with advanced pancreatic adenocarcinoma (PA) treated with FOLFIRINOX experience adverse events requiring dose reduction. We aimed to assess the association between relative dose intensity (RDI) and disease control in a European setting. Methods: We retrospectively included patients with advanced PA treated with three or more cycles of FOLFIRINOX between 2011 and 2018 in six French centers. We computed the cumulative single-agent RDI (csRDI) before the first reassessment for each FOLFIRINOX agent (oxaliplatin, irinotecan, 5FU bolus, and 5FU intravenous infusion) and the cumulative multi-drug RDI (cmRDI) of their combination. The association between RDI and disease control or objective response at first reassessment was evaluated using multivariable logistic regression models controlling for performance status, liver metastases, and center. Results: We included 243 patients. Median csRDIs were 81%, 79%, 75%, and 85% for oxaliplatin, irinotecan, 5FU bolus, and 5FU intravenous infusion, respectively. Median cmRDI was 80%. None of the RDIs was significantly associated with disease control or objective response. Including RDI in a clinical model did not improve its ability to predict disease control; the area under the curve was 0.79 (95% CI: 0.73–0.85) with RDI versus 0.78 (95% CI: 0.72–0.85) without. Similar results were observed for the objective response. Conclusion: Pragmatic dose adjustments of FOLFIRINOX should be made by oncologists without considering a loss of effect.
Collapse
Affiliation(s)
| | | | | | | | - Claire Cheymol
- Onco-Hematology Department, Saint Vincent de Paul Hospital, Lille, France
| | - Emilia Rad
- Medical Oncology Department, Victor Provo Hospital, Roubaix, France
| | - Farid El Hajbi
- Medical Oncology Department, Oscar Lambret Center, Lille, France
| | - Astrid Lièvre
- Department of Gastroenterology, CHU Pontchaillou, Rennes, France
| | - Julien Edeline
- Medical Oncology Department, Eugène Marquis Center, Rennes, France
| | | | | | - Anthony Turpin
- University of Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR-S 1277 - Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
11
|
Innominato PF, Karaboué A, Focan C, Chollet P, Giacchetti S, Bouchahda M, Ulusakarya A, Torsello A, Adam R, Lévi FA, Garufi C. Efficacy and safety of chronomodulated irinotecan, oxaliplatin, 5-fluorouracil and leucovorin combination as first- or second-line treatment against metastatic colorectal cancer: Results from the International EORTC 05011 Trial. Int J Cancer 2021; 148:2512-2521. [PMID: 33270911 PMCID: PMC8048520 DOI: 10.1002/ijc.33422] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/20/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
The triplet combination of irinotecan, oxaliplatin and fluorouracil is an active frontline regimen in metastatic colorectal cancer, but scarce data exist on its use as salvage treatment. We aimed at assessing its safety and efficacy profiles with its circadian-based administration (chronoIFLO5) as either first- or second-line treatment, within the time-finding EORTC 05011 trial. Five-day chronoIFLO5 was administered every 3 weeks in patients with PS 0, 1 or 2. It consisted of chronomodulated irinotecan (180 mg/sqm), oxaliplatin (80 mg/sqm) and fluorouracil-leucovorin (2800 and 1200 mg/sqm, respectively). For our study, toxicity and antitumour activity were evaluated separately in first- and second-line settings. Primary endpoints included Grade 3-4 toxicity rates, best objective response rate (ORR), progression-free survival (PFS) and overall survival (OS). One-hundred forty-nine and 44 patients were treated in first-line and second-line settings, respectively, with a total of 1138 cycles with median relative dose intensities of about 90%. Demographics were comparable in the two groups. Thirty-six (24.7%) and 10 (22.2%) patients experienced at least one episode of severe toxicity in first line and second line, respectively. Frontline chronoIFLO5 yielded an ORR of 62.3% [95% CI: 54.2-70.4] and resulted in median PFS and OS of 8.7 months [7.5-9.9] and 19.9 months [15.4-24.5]. Corresponding figures in second line were 37.5% [22.5-52.5], 6.7 months [4.8-8.9] and 16.3 months [11.8-20.8]. International and prospective evaluation revealed the favourable safety and efficacy profiles of chronoIFLO5, both as frontline and as salvage treatment against metastatic colorectal cancer. In particular, encouraging activity in second line was observed, with limited haematological toxicity.
Collapse
Affiliation(s)
- Pasquale F. Innominato
- North Wales Cancer Centre, Ysbyty Gwynedd, Betsi Cadwaladr University Health BoardBangorUK
- Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical SciencesWarwick Medical SchoolCoventryUK
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
| | - Abdoulaye Karaboué
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Medical Oncology UnitGHI Le Raincy‐MontfermeilMontfermeilFrance
| | - Christian Focan
- Department of OncologyCHC‐MontLegia, Groupe Santé CHC‐LiègeLiègeBelgium
| | - Philippe Chollet
- Clinical and Translational Research DivisionJean Perrin Comprehensive Cancer CentreClermont‐FerrandFrance
| | - Sylvie Giacchetti
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Department of OncologySaint Louis Hospital, Public Hospitals of Paris (AP‐HP)ParisFrance
| | - Mohamed Bouchahda
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Medical Oncology UnitClinique du MousseauEvryFrance
- Medical Oncology UnitClinique Saint Jean L'ErmitageMelunFrance
- Chronotherapy Unit, Department of Medical OncologyPaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Ayhan Ulusakarya
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Chronotherapy Unit, Department of Medical OncologyPaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Angela Torsello
- Division of Medical OncologySan Giovanni‐ Addolorata HospitalRomeItaly
| | - René Adam
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Hepatobiliary CentrePaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Francis A. Lévi
- Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical SciencesWarwick Medical SchoolCoventryUK
- UPR “Chronotherapy, Cancers and Transplantation”, Faculty of MedicineParis‐Saclay UniversityVillejuifFrance
- Hepatobiliary CentrePaul Brousse Hospital, Public Hospitals of Paris (AP‐HP)VillejuifFrance
| | - Carlo Garufi
- Division of Medical OncologySan Camillo Forlanini HospitalRomeItaly
| |
Collapse
|
12
|
Deyme L, Barbolosi D, Mbatchi LC, Tubiana-Mathieu N, Ychou M, Evrard A, Gattacceca F. Population pharmacokinetic model of irinotecan and its four main metabolites in patients treated with FOLFIRI or FOLFIRINOX regimen. Cancer Chemother Pharmacol 2021; 88:247-258. [PMID: 33912999 DOI: 10.1007/s00280-021-04255-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 03/07/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE The aim of the present study was to characterize the pharmacokinetics of irinotecan and its four main metabolites (SN-38, SN-38G, APC and NPC) in metastatic colorectal cancer patients treated with FOLFIRI and FOLFIRINOX regimens and to quantify and explain the inter-individual pharmacokinetic variability in this context. METHODS A multicenter study including 109 metastatic colorectal cancer patients treated with FOLFIRI or FOLFIRINOX regimen, associated or not with a monoclonal antibody, was conducted. Concentrations of irinotecan and its four main metabolites were measured in 506 blood samples during the first cycle of treatment. Collected data were analyzed using the population approach. First, fixed and random effects models were selected using statistical and graphical methods; second, the impact of covariates on pharmacokinetic parameters was evaluated to explain the inter-individual variability in pharmacokinetic parameters. RESULTS A seven-compartment model best described the pharmacokinetics of irinotecan and its four main metabolites. First-order rates were assigned to distribution, elimination, and metabolism processes, except for the transformation of irinotecan to NPC which was nonlinear. Addition of a direct conversion of NPC into SN-38 significantly improved the model. Co-administration of oxaliplatin significantly modified the distribution of SN-38. CONCLUSION To our knowledge, the present model is the first to allow a simultaneous description of irinotecan pharmacokinetics and of its four main metabolites. Moreover, a direct conversion of NPC into SN-38 had never been described before in a population pharmacokinetic model of irinotecan. The model will be useful to develop pharmacokinetic-pharmacodynamic models relating SN-38 concentrations to efficacy and digestive toxicities. CLINICAL TRIALS REGISTRATION NUMBER ClinicalTrials.gov identifier: NCT00559676.
Collapse
Affiliation(s)
- Laure Deyme
- SMARTc, Centre de Recherche en Cancérologie de Marseille (CRCM), Faculté de Pharmacie, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France.
| | - Dominique Barbolosi
- SMARTc, Centre de Recherche en Cancérologie de Marseille (CRCM), Faculté de Pharmacie, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| | - Litaty Céphanoée Mbatchi
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nîmes-Carémeau, Nîmes, France.,IRCM, Inserm U1194, Université de Montpellier, Montpellier, France
| | | | - Marc Ychou
- Institut Régional du Cancer de Montpellier (ICM)-Val d'Aurelle, Montpellier, France
| | - Alexandre Evrard
- Laboratoire de Biochimie et Biologie Moléculaire, CHU Nîmes-Carémeau, Nîmes, France.,IRCM, Inserm U1194, Université de Montpellier, Montpellier, France
| | - Florence Gattacceca
- SMARTc, Centre de Recherche en Cancérologie de Marseille (CRCM), Faculté de Pharmacie, INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
13
|
Minichmayr IK, Karlsson MO, Jönsson S. Pharmacometrics-Based Considerations for the Design of a Pharmacogenomic Clinical Trial Assessing Irinotecan Safety. Pharm Res 2021; 38:593-605. [PMID: 33733372 PMCID: PMC8057977 DOI: 10.1007/s11095-021-03024-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/26/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Pharmacometric models provide useful tools to aid the rational design of clinical trials. This study evaluates study design-, drug-, and patient-related features as well as analysis methods for their influence on the power to demonstrate a benefit of pharmacogenomics (PGx)-based dosing regarding myelotoxicity. METHODS Two pharmacokinetic and one myelosuppression model were assembled to predict concentrations of irinotecan and its metabolite SN-38 given different UGT1A1 genotypes (poor metabolizers: CLSN-38: -36%) and neutropenia following conventional versus PGx-based dosing (350 versus 245 mg/m2 (-30%)). Study power was assessed given diverse scenarios (n = 50-400 patients/arm, parallel/crossover, varying magnitude of CLSN-38, exposure-response relationship, inter-individual variability) and using model-based data analysis versus conventional statistical testing. RESULTS The magnitude of CLSN-38 reduction in poor metabolizers and the myelosuppressive potency of SN-38 markedly influenced the power to show a difference in grade 4 neutropenia (<0.5·109 cells/L) after PGx-based versus standard dosing. To achieve >80% power with traditional statistical analysis (χ2/McNemar's test, α = 0.05), 220/100 patients per treatment arm/sequence (parallel/crossover study) were required. The model-based analysis resulted in considerably smaller total sample sizes (n = 100/15 given parallel/crossover design) to obtain the same statistical power. CONCLUSIONS The presented findings may help to avoid unfeasible trials and to rationalize the design of pharmacogenetic studies.
Collapse
Affiliation(s)
- Iris K Minichmayr
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Siv Jönsson
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden.
| |
Collapse
|
14
|
Schneider JJ, Galettis P, Martin JH. Overcoming barriers to implementing precision dosing with 5-fluorouracil and capecitabine. Br J Clin Pharmacol 2021; 87:317-325. [PMID: 33386659 DOI: 10.1111/bcp.14723] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/01/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Despite advances in targeted cancer therapy, the fluoropyrimidines 5-fluorouracil (5FU) and capecitabine continue to play an important role in oncology. Historically, dosing of these drugs has been based on body surface area. This approach has been demonstrated to be an imprecise way to determine the optimal dose for a patient. Evidence in the literature has demonstrated that precision dosing approaches, such as DPD enzyme activity testing and, in the case of intravenous 5FU, pharmacokinetic-guided dosing, can reduce toxicity and yield better patient outcomes. However, despite the evidence, there has not been uniform adoption of these approaches in the clinical setting. When a drug such as 5FU has been used clinically for many decades, it may be difficult to change clinical practice. With the aim of facilitating change of practice, issues and barriers to implementing precision dosing approaches for 5FU and capecitabine are identified and discussed with possible solutions proposed.
Collapse
Affiliation(s)
- Jennifer J Schneider
- Discipline of Clinical Pharmacology, School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia.,Centre for Drug Repurposing and Medicines Research, Level 3 Hunter Medical Research Institute, Kookaburra Circuit, Newcastle, New South Wales, Australia
| | - Peter Galettis
- Discipline of Clinical Pharmacology, School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia.,Centre for Drug Repurposing and Medicines Research, Level 3 Hunter Medical Research Institute, Kookaburra Circuit, Newcastle, New South Wales, Australia
| | - Jennifer H Martin
- Discipline of Clinical Pharmacology, School of Medicine and Public Health, University of Newcastle, Newcastle, New South Wales, Australia.,Centre for Drug Repurposing and Medicines Research, Level 3 Hunter Medical Research Institute, Kookaburra Circuit, Newcastle, New South Wales, Australia
| |
Collapse
|
15
|
Godinho PAR, Silva PGB, Lisboa MRP, Costa BA, Gifoni MAC, Rocha Filho DR, Lima-Júnior RCP, Vale ML. Electronic von Frey as an objective assessment tool for oxaliplatin-induced peripheral neuropathy: A prospective longitudinal study. Eur J Cancer Care (Engl) 2020; 30:e13360. [PMID: 33219575 DOI: 10.1111/ecc.13360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/28/2020] [Accepted: 10/15/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVE There is wide discrepancy on how to perform clinical assessment of oxaliplatin-induced peripheral neuropathy. In this scenario, the Electronic von Frey (EVF), which evaluates pain objectively based upon mechanical pain thresholds (MPTs), may be a valuable tool. The present study aims to quantify hyperalgesia in the hands and feet of patients treated with oxaliplatin and to propose a novel method to classify the degree of neurotoxicity using EVF-derived measures as cut-off points. METHODS This is a prospective cohort study including 46 patients treated for colorectal cancer with the FLOX regimen. Before each oxaliplatin administration, patients were evaluated with the Acute and Chronic Neuropathy Questionnaire, Oxaliplatin-Specific Neurotoxicity Scale and National Cancer Institute Common Terminology Criteria for Adverse Events scale. Also, objective pain assessment with the EVF was performed. RESULTS For both upper and lower extremities, EVF was shown to correlate well with patients' symptoms and functional impairment, as assessed by subjective scales. Also, when cut-off MPT variations were determined for diagnosis of neurotoxicity grade 2 or 3, the method showed good sensitivity and specificity. CONCLUSION Electronic von Frey is a noninvasive and easy-to-perform objective method with potential to supplement the current assessment tools for oxaliplatin-induced peripheral neuropathy, which are mostly subjective.
Collapse
Affiliation(s)
- Priscilla A R Godinho
- Graduate Program in Pharmacology, School of Medicine, Federal University of Ceará (UFC, Fortaleza, Brazil
| | - Paulo G B Silva
- Centro Universitário Christus (UNICHRISTUS, Fortaleza, Brazil
| | - Mario R P Lisboa
- Centro Universitário Christus (UNICHRISTUS, Fortaleza, Brazil.,Department of Morphology, Faculty of Medicine, Universidade Federal do Ceará (UFC, Fortaleza, Brazil
| | - Bruno A Costa
- Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará (UFC, Fortaleza, Brazil
| | - Markus A C Gifoni
- Clinical Oncology Department, Instituto do Câncer do Ceará (ICC, Fortaleza, Brazil
| | - Duílio R Rocha Filho
- Clinical Oncology Department, Instituto do Câncer do Ceará (ICC, Fortaleza, Brazil
| | - Roberto C P Lima-Júnior
- Graduate Program in Pharmacology, School of Medicine, Federal University of Ceará (UFC, Fortaleza, Brazil.,Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará (UFC, Fortaleza, Brazil
| | - Mariana L Vale
- Graduate Program in Pharmacology, School of Medicine, Federal University of Ceará (UFC, Fortaleza, Brazil.,Department of Morphology, Faculty of Medicine, Universidade Federal do Ceará (UFC, Fortaleza, Brazil.,Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará (UFC, Fortaleza, Brazil
| |
Collapse
|
16
|
Moser T, Waldispuehl-Geigl J, Belic J, Weber S, Zhou Q, Hasenleithner SO, Graf R, Terzic JA, Posch F, Sill H, Lax S, Kashofer K, Hoefler G, Schoellnast H, Heitzer E, Geigl JB, Bauernhofer T, Speicher MR. On-treatment measurements of circulating tumor DNA during FOLFOX therapy in patients with colorectal cancer. NPJ Precis Oncol 2020; 4:30. [PMID: 33299124 PMCID: PMC7666126 DOI: 10.1038/s41698-020-00134-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/21/2020] [Indexed: 01/20/2023] Open
Abstract
We addressed a significant unknown feature of circulating tumor DNA (ctDNA), i.e., how ctDNA levels change during chemotherapy, by serially monitoring ctDNA in patients with colorectal cancer during the 48-h application of FOLFOX. Surprisingly, we did not observe a spike in ctDNA as a sign of a responsive tumor, but instead ctDNA levels initially decreased and remained low in patients with stable disease or partial response. Our observations reveal further insights into cell destruction during chemotherapy with important implications for the management of patients.
Collapse
Affiliation(s)
- Tina Moser
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Julie Waldispuehl-Geigl
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Jelena Belic
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
- Cancer Research UK Cambridge Institute, University of Cambridge, CB2 0RE, Cambridge, UK
| | - Sabrina Weber
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Qing Zhou
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Samantha O Hasenleithner
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Ricarda Graf
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Jasmin Alia Terzic
- Department of Internal Medicine Graz, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Department of Internal Medicine Graz, Division of Oncology, Medical University of Graz, Graz, Austria
| | - Heinz Sill
- Department of Internal Medicine, Division of Hematology, Medical University of Graz, Graz, Austria
| | - Sigurd Lax
- Department of Pathology, General Hospital Graz II, Graz, Austria, and Johannes Kepler University Linz, Linz, Austria
| | - Karl Kashofer
- Institute of Pathology, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Gerald Hoefler
- Institute of Pathology, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Helmut Schoellnast
- Department of Radiology, Division of General Radiology, Medical University of Graz, Graz, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Christian Doppler Laboratory for Liquid Biopsies for Early Detection of Cancer, Graz, Austria
| | - Jochen B Geigl
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria
| | - Thomas Bauernhofer
- Department of Internal Medicine Graz, Division of Oncology, Medical University of Graz, Graz, Austria.
| | - Michael R Speicher
- Institute of Human Genetics, Diagnostic and Research Center for Molecular BioMedicine, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
17
|
Maillard M, Le Louedec F, Thomas F, Chatelut E. Diversity of dose-individualization and therapeutic drug monitoring practices of platinum compounds: a review. Expert Opin Drug Metab Toxicol 2020; 16:907-925. [PMID: 33016786 DOI: 10.1080/17425255.2020.1789590] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Platinum-derived drugs are commonly used for the treatment of solid tumors. The differences in chemical structures of these molecules lead to different pharmacological properties, in terms of indication, efficacy, and toxicity. Their pharmacokinetics (PK) differ according to their respective renal elimination and have led to many studies investigating their dose optimization. Area covered: This review attempts to summarize and compare PK and pharmacodynamics of cisplatin, carboplatin, and oxaliplatin, with an emphasis on differences of dose calculations and opportunities for therapeutic drug monitoring (TDM) in various patient populations. Expert opinion: Although cisplatin and carboplatin can be considered as analogs since they share the same DNA interacting properties, the slower hydrolysis of the latter results in a better safety profile. Carboplatin is the only drug in oncology to be administrated according to a target area under the curve of concentration versus time, considering that its PK variability is almost fully explained by renal function, not by body size. This enables individual dosing based on predicted carboplatin clearance (along with patients renal characteristics) or on actual clearance with TDM, especially in a high-dose protocol.
Collapse
Affiliation(s)
- Maud Maillard
- Laboratoire De Pharmacologie, Institut Claudius-Regaud, IUCT-Oncopole , Toulouse Cedex 9, France.,Cancer Research Center of Toulouse, INSERM UMR1037, Team 14 DIAD (Dose Individualization of Anticancer Drug) , Toulouse, France.,Faculté de Pharmacie, Université Paul Sabatier Toulouse III , Toulouse, France
| | - Félicien Le Louedec
- Laboratoire De Pharmacologie, Institut Claudius-Regaud, IUCT-Oncopole , Toulouse Cedex 9, France.,Cancer Research Center of Toulouse, INSERM UMR1037, Team 14 DIAD (Dose Individualization of Anticancer Drug) , Toulouse, France.,Faculté de Pharmacie, Université Paul Sabatier Toulouse III , Toulouse, France
| | - Fabienne Thomas
- Laboratoire De Pharmacologie, Institut Claudius-Regaud, IUCT-Oncopole , Toulouse Cedex 9, France.,Cancer Research Center of Toulouse, INSERM UMR1037, Team 14 DIAD (Dose Individualization of Anticancer Drug) , Toulouse, France.,Faculté de Pharmacie, Université Paul Sabatier Toulouse III , Toulouse, France
| | - Etienne Chatelut
- Laboratoire De Pharmacologie, Institut Claudius-Regaud, IUCT-Oncopole , Toulouse Cedex 9, France.,Cancer Research Center of Toulouse, INSERM UMR1037, Team 14 DIAD (Dose Individualization of Anticancer Drug) , Toulouse, France.,Faculté de Pharmacie, Université Paul Sabatier Toulouse III , Toulouse, France
| |
Collapse
|
18
|
Teng C, Reuter SE, Blinman PL, Dhillon HM, Galettis P, Proschogo N, McLachlan AJ, Vardy JL. Ibudilast for prevention of oxaliplatin-induced acute neurotoxicity: a pilot study assessing preliminary efficacy, tolerability and pharmacokinetic interactions in patients with metastatic gastrointestinal cancer. Cancer Chemother Pharmacol 2020; 86:547-558. [PMID: 32949265 DOI: 10.1007/s00280-020-04143-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/06/2020] [Indexed: 01/07/2023]
Abstract
PURPOSE This prospective, open-label, sequential 'before vs. after' pilot study was conducted to provide preliminary efficacy and tolerability data for ibudilast in the prevention of oxaliplatin-induced neurotoxicity in patients with metastatic upper gastrointestinal or colorectal cancer. Any potential impact of ibudilast on oxaliplatin and 5-fluorouracil pharmacokinetics was also explored. METHODS Participants were administered a chemotherapy cycle (FOLFOX or CapeOx), followed by a chemotherapy cycle with co-administration of ibudilast 30 mg b.i.d. p.o. Efficacy was assessed on Day 3 and end of cycle using the Oxaliplatin-Specific Neurotoxicity Scale (OSNS) and additional clinical/patient-reported neurotoxicity measures. A population pharmacokinetic approach was used to determine oxaliplatin and 5-fluorouracil pharmacokinetics with and without ibudilast. RESULTS Sixteen participants consented; 14 completed both chemotherapy cycles. Across all measures, the majority of participants experienced either an improvement or no worsening of neurotoxicity with ibudilast treatment. Based on OSNS assessments, acute neurotoxicity was unchanged in 12/14 participants and improved in 2/14 participants. The 90% confidence interval (CI) of the dose-normalised ratio of oxaliplatin AUC (90% CI 95.0-109%) and 5-fluorouracil AUC (90% CI 66.5-173%) indicated no significant impact of ibudilast on systemic exposure. CONCLUSION This pilot study indicated ibudilast co-administration may improve or stabilise oxaliplatin-induced neurotoxicity. Given the expected worsening of symptoms in patients with continued chemotherapy, this represents a signal of effect that warrants further investigation. Pharmacokinetic analysis indicates ibudilast has no significant effect on oxaliplatin pharmacokinetics, and is unlikely to influence pharmacokinetics of 5-fluorouracil. CLINICAL TRIAL REGISTRATION Trial registration number: UTN U1111-1209-0075 and ANZCTRN12618000232235 (registered 13/02/2018).
Collapse
Affiliation(s)
- Christina Teng
- Department of Medical Oncology, Concord Cancer Centre, Concord Repatriation General Hospital, Concord, Australia
- Central Coast Cancer Centre, Gosford, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Stephanie E Reuter
- Clinical and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Prunella L Blinman
- Department of Medical Oncology, Concord Cancer Centre, Concord Repatriation General Hospital, Concord, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia
| | - Haryana M Dhillon
- Centre for Medical Psychology and Evidence-Based Decision-Making, University of Sydney, Camperdown, Australia
| | - Peter Galettis
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | | | | | - Janette L Vardy
- Department of Medical Oncology, Concord Cancer Centre, Concord Repatriation General Hospital, Concord, Australia.
- Centre for Medical Psychology and Evidence-Based Decision-Making, University of Sydney, Camperdown, Australia.
- Faculty of Medicine and Health, The University of Sydney, Camperdown, Australia.
| |
Collapse
|
19
|
Macaire P, Paris J, Vincent J, Ghiringhelli F, Bengrine-Lefevre L, Schmitt A. Impact of granulocyte colony-stimulating factor on FOLFIRINOX-induced neutropenia prevention: A population pharmacokinetic/pharmacodynamic approach. Br J Clin Pharmacol 2020; 86:2473-2485. [PMID: 32386071 DOI: 10.1111/bcp.14356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/21/2020] [Accepted: 04/30/2020] [Indexed: 12/18/2022] Open
Abstract
AIMS Granulocyte colony-stimulating factor (G-CSF) is frequently prescribed to prevent chemotherapy-induced neutropenia, but the administration schedule remains empirical in case of bimonthly chemotherapy such as FOLFIRINOX regimen. This pharmacokinetic/pharmacodynamic (PK/PD) study was performed to determine the effect of different G-CSF regimens on the incidence and duration of neutropenia following FOLFIRINOX administration in order to propose an optimal G-CSF dosing schedule. METHODS A population PK/PD model was developed to describe individual neutrophil time course from absolute neutrophil counts (ANC) obtained in 40 advanced cancer patients receiving FOLFIRINOX regimen. The structural model considered ANC dynamics, neutropenic effect of cytotoxics and the stimulating effect of G-CSF on neutrophils. Final model estimates were used to simulate different G-CSF dosing schedules for 1000 virtual subjects. The incidence and duration of neutropenia were then calculated for different G-CSF dosing schedules. RESULTS The final model successfully described the myelosuppressive effect induced by the 3 cytotoxics for all patients. Simulations showed that pegfilgrastim administration reduced the risk of severe neutropenia by 22.9% for subjects with low ANC at the start of chemotherapy. Median duration in this group was also shortened by 3.1 days when compared to absence of G-CSF. Delayed G-CSF administration was responsible for higher incidence and longer duration of neutropenia compared to absence of administration. CONCLUSION The PK/PD model well described our population's ANC data. Simulations showed that pegylated-G-CSF administration 24 hours after the end of chemotherapy seems to be the optimal schedule to reduce FOLFIRINOX-induced neutropenia. We also underline the potential negative effect of G-CSF maladministration.
Collapse
Affiliation(s)
- Pauline Macaire
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| | - Justine Paris
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| | - Julie Vincent
- Oncology Department, Centre Georges-François Leclerc, Dijon, France
| | - François Ghiringhelli
- INSERM U1231, University of Burgundy Franche-Comté, Dijon, France.,Oncology Department, Centre Georges-François Leclerc, Dijon, France
| | | | - Antonin Schmitt
- Pharmacy Department, Centre Georges-François Leclerc, Dijon, France.,INSERM U1231, University of Burgundy Franche-Comté, Dijon, France
| |
Collapse
|
20
|
Arshad U, Ploylearmsaeng SA, Karlsson MO, Doroshyenko O, Langer D, Schömig E, Kunze S, Güner SA, Skripnichenko R, Ullah S, Jaehde U, Fuhr U, Jetter A, Taubert M. Prediction of exposure-driven myelotoxicity of continuous infusion 5-fluorouracil by a semi-physiological pharmacokinetic-pharmacodynamic model in gastrointestinal cancer patients. Cancer Chemother Pharmacol 2020; 85:711-722. [PMID: 32152679 PMCID: PMC7125253 DOI: 10.1007/s00280-019-04028-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/31/2019] [Indexed: 01/07/2023]
Abstract
Purpose To describe 5-fluorouracil (5FU) pharmacokinetics, myelotoxicity and respective covariates using a simultaneous nonlinear mixed effect modelling approach. Methods Thirty patients with gastrointestinal cancer received 5FU 650 or 1000 mg/m2/day as 5-day continuous venous infusion (14 of whom also received cisplatin 20 mg/m2/day). 5FU and 5-fluoro-5,6-dihydrouracil (5FUH2) plasma concentrations were described by a pharmacokinetic model using NONMEM. Absolute leukocyte counts were described by a semi-mechanistic myelosuppression model. Covariate relationships were evaluated to explain the possible sources of variability in 5FU pharmacokinetics and pharmacodynamics. Results Total clearance of 5FU correlated with body surface area (BSA). Population estimate for total clearance was 249 L/h. Clearances of 5FU and 5FUH2 fractionally changed by 77%/m2 difference from the median BSA. 5FU central and peripheral volumes of distribution were 5.56 L and 28.5 L, respectively. Estimated 5FUH2 clearance and volume of distribution were 121 L/h and 96.7 L, respectively. Baseline leukocyte count of 6.86 × 109/L, as well as mean leukocyte transit time of 281 h accounting for time delay between proliferating and circulating cells, was estimated. The relationship between 5FU plasma concentrations and absolute leukocyte count was found to be linear. A higher degree of myelosuppression was attributed to combination therapy (slope = 2.82 L/mg) with cisplatin as compared to 5FU monotherapy (slope = 1.17 L/mg). Conclusions BSA should be taken into account for predicting 5FU exposure. Myelosuppression was influenced by 5FU exposure and concomitant administration of cisplatin. Electronic supplementary material The online version of this article (10.1007/s00280-019-04028-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Usman Arshad
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany.
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany.
| | - Su-Arpa Ploylearmsaeng
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Oxana Doroshyenko
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Dorothee Langer
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Edgar Schömig
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Sabine Kunze
- Department of Radiotherapy, University Hospital Cologne, Cologne, Germany
| | - Semih A Güner
- Department of Radiotherapy, University Hospital Cologne, Cologne, Germany
| | | | - Sami Ullah
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Ulrich Jaehde
- Institute of Pharmacy, Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Uwe Fuhr
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| | - Alexander Jetter
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Max Taubert
- Faculty of Medicine and University Hospital Cologne, Center for Pharmacology, Department I of Pharmacology, University of Cologne, Cologne, Germany
| |
Collapse
|
21
|
Hill RJW, Innominato PF, Lévi F, Ballesta A. Optimizing circadian drug infusion schedules towards personalized cancer chronotherapy. PLoS Comput Biol 2020; 16:e1007218. [PMID: 31986133 PMCID: PMC7004559 DOI: 10.1371/journal.pcbi.1007218] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 02/06/2020] [Accepted: 11/21/2019] [Indexed: 11/18/2022] Open
Abstract
Precision medicine requires accurate technologies for drug administration and proper systems pharmacology approaches for patient data analysis. Here, plasma pharmacokinetics (PK) data of the OPTILIV trial in which cancer patients received oxaliplatin, 5-fluorouracil and irinotecan via chronomodulated schedules delivered by an infusion pump into the hepatic artery were mathematically investigated. A pump-to-patient model was designed in order to accurately represent the drug solution dynamics from the pump to the patient blood. It was connected to semi-mechanistic PK models to analyse inter-patient variability in PK parameters. Large time delays of up to 1h41 between the actual pump start and the time of drug detection in patient blood was predicted by the model and confirmed by PK data. Sudden delivery spike in the patient artery due to glucose rinse after drug administration accounted for up to 10.7% of the total drug dose. New model-guided delivery profiles were designed to precisely lead to the drug exposure intended by clinicians. Next, the complete mathematical framework achieved a very good fit to individual time-concentration PK profiles and concluded that inter-subject differences in PK parameters was the lowest for irinotecan, intermediate for oxaliplatin and the largest for 5-fluorouracil. Clustering patients according to their PK parameter values revealed patient subgroups for each drug in which inter-patient variability was largely decreased compared to that in the total population. This study provides a complete mathematical framework to optimize drug infusion pumps and inform on inter-patient PK variability, a step towards precise and personalized cancer chronotherapy. Accuracy and safety of infusion pumps remain a critical issue in the clinics and the development of accurate mathematical models to optimize drug administration though such devices has a key part to play in the advancement of precision medicine. Here, PK data from cancer patient receiving irinotecan, oxaliplatin and 5-fluorouracil into the hepatic artery via an infusion pump was mathematically investigated. A pump-to-patient model was designed and revealed significant inconsistencies between intended drug profiles and actual plasma concentrations. This mathematical model was then used to suggest improved profiles in order to minimise error and optimise delivery. Physiologically-based PK models of the three drugs were then linked to the pump-to-patient model. The whole framework achieved a very good fit to data and allowed quantifying inter-patient variability in PK parameters and linking them to potential clinical biomarkers via patient clustering. The developed methodology improves our understanding of patient-specific drug pharmacokinetics towards personalized drug administration.
Collapse
Affiliation(s)
- Roger J W Hill
- EPSRC & MRC Centre for Doctoral Training in Mathematics for Real-World Systems, University of Warwick, Coventry, UK
| | - Pasquale F Innominato
- North Wales Cancer Centre, Ysbyty Gwynedd, Betsi Cadwaladr University Health Board, Bangor, UK.,Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical Sciences, Warwick Medical School, Coventry, UK
| | - Francis Lévi
- Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical Sciences, Warwick Medical School, Coventry, UK.,INSERM and Paris Sud university, UMRS 935, Team "Cancer Chronotherapy and Postoperative Liver Functions", Campus CNRS, Villejuif, F-94807, France. & Honorary position, University of Warwick, UK
| | - Annabelle Ballesta
- INSERM and Paris Sud university, UMRS 935, Team "Cancer Chronotherapy and Postoperative Liver Functions", Campus CNRS, Villejuif, F-94807, France. & Honorary position, University of Warwick, UK
| |
Collapse
|
22
|
Sugarman R, Patel R, Sharma S, Plenker D, Tuveson D, Saif MW. Pharmacokinetics and pharmacodynamics of new drugs for pancreatic cancer. Expert Opin Drug Metab Toxicol 2019; 15:541-552. [PMID: 31241371 DOI: 10.1080/17425255.2019.1637417] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: Pancreatic cancer (PC) remains a disease with a dismal prognosis. Despite accounting for only 3% of cancer diagnosis, 7% of all cancer deaths in the United States are from PC. This is explained by many being diagnosed with late-stage disease and the cancer's resistance to chemotherapy. Since 1996 there have only been two upfront regimens found to be superior to gemcitabine, FOLFIRINOX (5-fluorouracil/leucovorin and oxaliplatin) and gemcitabine plus nab-paclitaxel. Areas covered: Clinical pharmacology of newer agents that are either approved or being investigated in the management of PC. Knowledge of their pharmacokinetics, pharmacodynamics, and pharmacogenetics can be used to predict outcomes for specific patient populations. Drugs discussed include nanoliposomal irinotecan, pegvorhyaluronidase alfa, poly (ADP-ribose) polymerase enzyme inhibitors, larotrectinib, and napabucasin. Expert opinion: PC is a heterogeneous disease and outcomes are likely to improve as better predictive models of an individual's response to different therapies are developed. This may be best accomplished through phase 0 studies and the use of tumor organoid models grown from initial biopsies or resected tissue. The genetic and physical makeup of the tumor as well as the functional characterization in patient-derived organoids (PDOs), can help guide which agents may be most efficacious or toxic.
Collapse
Affiliation(s)
- Ryan Sugarman
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| | - Rajvi Patel
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| | - Sandhya Sharma
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| | - Dennis Plenker
- b Cold Spring Harbor Laboratory , Cold Spring Harbor , NY , USA
| | - David Tuveson
- b Cold Spring Harbor Laboratory , Cold Spring Harbor , NY , USA
| | - Muhammad Wasif Saif
- a Northwell Health Cancer Institute , Donald and Barbara Zucker School of Medicine at Hofstra/Northwell , Lake Success , NY , USA
| |
Collapse
|