1
|
Kyriacou NM, Gross AS, McLachlan AJ. Inter-Ethnic Differences in the Efficacy and Safety of Tyrosine Kinase Inhibitors Used in Oncology: Insights From Phase 3 Clinical Trials. Clin Transl Sci 2025; 18:e70224. [PMID: 40296413 PMCID: PMC12037692 DOI: 10.1111/cts.70224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Differences in the efficacy and safety of tyrosine kinase inhibitors (TKIs) have been observed across ethnic/ancestry subpopulations (previously reviewed to 2017). With an expanding number of TKIs approved since that time, an updated review of TKI response across ethnic/ancestry subpopulations in Phase 3 TKI clinical trials was conducted. A total of 73 population subgroup analyses (defined by participant race, ethnicity, ancestry or geographic region) of progression-free survival (PFS) and/or overall survival (OS) were identified by a literature search. Twelve (16%) of the analyses investigating the efficacy of afatinib, brigatinib, dacomitinib, gilteritinib, lorlatinib, neratinib, osimertinib, or pazopanib were assessed to report population differences in PFS and/or OS. For 28 (38%) of the analyses that showed suggestions of a potential efficacy difference across subpopulations, limitations in the data available precluded further assessment. There were 17 (23%) analyses assessed to report comparable efficacy outcomes across diverse subpopulations. The majority of clinical trials noted no clinically remarkable differences in safety between subpopulations; however, for brigatinib, crizotinib, pazopanib, and sunitinib, distinct patterns of adverse events were reported in the Asian and non-Asian subgroups. The underrepresentation of specific subpopulations, the grouping together of results of diverse subpopulations, as well as inconsistencies in the definition and reporting of participant ethnicity/ancestry are barriers to the meaningful exploration of inter-ethnic differences in TKI response. Therefore, further insight into the associations between ethnicity/ancestry and TKI response will require an increase in the diversity of clinical trial participants and appropriate analysis and reporting of subpopulation results.
Collapse
Affiliation(s)
- Nicki M. Kyriacou
- Sydney Pharmacy School, Faculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Annette S. Gross
- Sydney Pharmacy School, Faculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Andrew J. McLachlan
- Sydney Pharmacy School, Faculty of Medicine and HealthUniversity of SydneySydneyAustralia
| |
Collapse
|
2
|
Buckle GC, DeBoer R, Xu MJ, Mrema A, Rubagumya F, Velloza J, Falade AS, Van Loon K. Using context-specific evidence to inform resource-stratified cancer guidelines: A call for a new approach. Cancer 2025; 131:e35573. [PMID: 39306723 PMCID: PMC11694159 DOI: 10.1002/cncr.35573] [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: 01/03/2025]
Abstract
Clinical practice guidelines are widely used in oncology to guide clinical decision making and inform health policy and planning. In recent years, the National Comprehensive Cancer Network and the American Society of Clinical Oncology, as well as other international groups, have developed resource-stratified guidelines to guide clinicians and policymakers on cancer diagnosis and management in settings with various levels of resource constraints. Current methods for developing resource-stratified guidelines rely heavily on supporting evidence originating from high-income countries. In this commentary, the authors discuss limitations of the existing methods to develop resource-stratified guidelines and offer perspective on ways to strengthen the guidelines and their evidence base. Pulling from conceptual frameworks in the health policy domain, the authors outline a more inclusive approach to evidence synthesis that seeks to integrate the growing volume of cancer research emerging from low- and middle-income countries. The authors also introduce a revised evidence framework that provides transparency into the generalizability of evidence within the guidelines. These changes have the potential to enhance resource-stratified guidelines and bring us one step closer to the goal of evidence-based guidelines that are appropriate for diverse settings and unique patient populations across the world.
Collapse
Affiliation(s)
- Geoffrey C. Buckle
- Division of Hematology/OncologyDepartment of MedicineUniversity of California San Francisco (UCSF)San FranciscoCaliforniaUSA
- Global Cancer ProgramUCSF Helen Diller Family Comprehensive Cancer CenterSan FranciscoCaliforniaUSA
| | - Rebecca DeBoer
- Division of Hematology/OncologyDepartment of MedicineUniversity of California San Francisco (UCSF)San FranciscoCaliforniaUSA
- Global Cancer ProgramUCSF Helen Diller Family Comprehensive Cancer CenterSan FranciscoCaliforniaUSA
| | - Mary Jue Xu
- Global Cancer ProgramUCSF Helen Diller Family Comprehensive Cancer CenterSan FranciscoCaliforniaUSA
- Department of Otolaryngology‐Head and Neck SurgeryUCSFSan FranciscoCaliforniaUSA
| | - Alita Mrema
- Ocean Road Cancer InstituteDar es SalaamTanzania
| | | | - Jennifer Velloza
- Department of Epidemiology and BiostatisticsUCSFSan FranciscoCaliforniaUSA
| | - Ayo S. Falade
- Mass General Brigham Hospital SalemSalemMassachusettsUSA
| | - Katherine Van Loon
- Division of Hematology/OncologyDepartment of MedicineUniversity of California San Francisco (UCSF)San FranciscoCaliforniaUSA
- Global Cancer ProgramUCSF Helen Diller Family Comprehensive Cancer CenterSan FranciscoCaliforniaUSA
| |
Collapse
|
3
|
Tominaga S, Yoshioka H, Hasegawa T, Suzui M, Maeda T, Miura N. Diurnal variation of cisplatin-induced renal toxicity in ICR mice. Biochem Biophys Res Commun 2024; 725:150266. [PMID: 38878759 DOI: 10.1016/j.bbrc.2024.150266] [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: 06/10/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/06/2024]
Abstract
Cisplatin (CDDP) is a platinum-based anticancer drug widely prescribed for its effectiveness in treating various forms of cancer. However, its major side effect is nephrotoxicity. Although several methods have been developed to mitigate CDDP-induced nephrotoxicity, an optimal approach has yet to be established. This study aimed to investigate the "chronotoxicity" of CDDP as a potential strategy to reduce its side effects. Male ICR mice were treated with CDDP (20 mg/kg, intraperitoneal injection, one shot) at zeitgeber time (ZT) 2 or ZT14 (light or dark phase). After 72 h, we collected plasma and kidney and evaluated several markers. We found that body weight change between ZT2 and ZT14 by CDDP was comparable. In contrast, many toxicological factors, such as plasma blood urine nitrogen, plasma creatinine, renal oxidative stress (malondialdehyde), DNA damage (γH2AX), acute kidney injury biomarker (KIM-1), and inflammation (Tnfα), were significantly induced at ZT14 compared to than that of ZT2. Our present data suggested that chronotoxicology might provide beneficial information on the importance of administration timings for toxic evaluations and unacceptable side effects.
Collapse
Affiliation(s)
- Sarah Tominaga
- Graduate School of Pharmaceutical Sciences, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan; Department of Neurotoxicology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Hiroki Yoshioka
- Faculty of Pharmacy, Gifu University of Medical Science, 4-3-3 Nijigaoka, Kani, Gifu, 509-0293, Japan; Department of Hygiene, Kitasato University, School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan; College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan.
| | - Tatsuya Hasegawa
- Department of Environmental Biochemistry, Mount Fuji Research Institute, 5597-1 Kamiyoshidakenmarubi, Fujiyoshida, Yamanashi, 403-0005, Japan
| | - Masumi Suzui
- Department of Neurotoxicology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Tohru Maeda
- Graduate School of Pharmaceutical Sciences, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan; College of Pharmacy, Kinjo Gakuin University, 2-1723 Omori, Moriyamaku, Nagoya, Aichi, 463-8521, Japan
| | - Nobuhiko Miura
- Department of Health Science, Yokohama University of Pharmacy, 601 Matano-cho, Totsuka-ku, Yokohama, Kanagawa, 245-2006, Japan.
| |
Collapse
|
4
|
DeRidder LB, Hare KA, Lopes A, Jenkins J, Fitzgerald N, MacPherson E, Fabian N, Morimoto J, Chu JN, Kirtane AR, Madani W, Ishida K, Kuosmanen JLP, Zecharias N, Colangelo CM, Huang HW, Chilekwa M, Lal NB, Srinivasan SS, Hayward AM, Wolpin BM, Trumper D, Quast T, Rubinson DA, Langer R, Traverso G. Closed-loop automated drug infusion regulator: A clinically translatable, closed-loop drug delivery system for personalized drug dosing. MED 2024; 5:780-796.e10. [PMID: 38663403 DOI: 10.1016/j.medj.2024.03.020] [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: 10/27/2023] [Revised: 01/26/2024] [Accepted: 03/21/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Dosing of chemotherapies is often calculated according to the weight and/or height of the patient or equations derived from these, such as body surface area (BSA). Such calculations fail to capture intra- and interindividual pharmacokinetic variation, which can lead to order of magnitude variations in systemic chemotherapy levels and thus under- or overdosing of patients. METHODS We designed and developed a closed-loop drug delivery system that can dynamically adjust its infusion rate to the patient to reach and maintain the drug's target concentration, regardless of a patient's pharmacokinetics (PK). FINDINGS We demonstrate that closed-loop automated drug infusion regulator (CLAUDIA) can control the concentration of 5-fluorouracil (5-FU) in rabbits according to a range of concentration-time profiles (which could be useful in chronomodulated chemotherapy) and over a range of PK conditions that mimic the PK variability observed clinically. In one set of experiments, BSA-based dosing resulted in a concentration 7 times above the target range, while CLAUDIA keeps the concentration of 5-FU in or near the targeted range. Further, we demonstrate that CLAUDIA is cost effective compared to BSA-based dosing. CONCLUSIONS We anticipate that CLAUDIA could be rapidly translated to the clinic to enable physicians to control the plasma concentration of chemotherapy in their patients. FUNDING This work was supported by MIT's Karl van Tassel (1925) Career Development Professorship and Department of Mechanical Engineering and the Bridge Project, a partnership between the Koch Institute for Integrative Cancer Research at MIT and the Dana-Farber/Harvard Cancer Center.
Collapse
Affiliation(s)
- Louis B DeRidder
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kyle A Hare
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aaron Lopes
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Josh Jenkins
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nina Fitzgerald
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emmeline MacPherson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Niora Fabian
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Josh Morimoto
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jacqueline N Chu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard Medical School, Boston, MA 02115, USA; Division of Gastroenterology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ameya R Kirtane
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wiam Madani
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Keiko Ishida
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Johannes L P Kuosmanen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Naomi Zecharias
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Hen-Wei Huang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Makaya Chilekwa
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nikhil B Lal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; MIT Media Lab, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shriya S Srinivasan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alison M Hayward
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brian M Wolpin
- Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David Trumper
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Troy Quast
- College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Douglas A Rubinson
- Harvard Medical School, Boston, MA 02115, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Giovanni Traverso
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
5
|
Patil N, Ma N, Mair M, Nazareth J, Sim A, Reynolds C, Freeman N, Chauhan M, Howells L, Peel D, Ahmad S, Sridhar T, Walter HS. Oral Cavity Cancers: Ethnic Differences in Radiotherapy Outcomes in a Majority South Asian Leicester Community. Clin Oncol (R Coll Radiol) 2024; 36:300-306. [PMID: 38388251 DOI: 10.1016/j.clon.2024.02.010] [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: 09/11/2023] [Revised: 01/15/2024] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
AIMS Squamous cell carcinoma oral cavity cancers (SCCOCCs) have a higher reported incidence in South Asian countries. We sought to compare presenting stage and outcome by ethnicity in patients with SCCOCC treated with radical radiotherapy in a single centre in the UK. MATERIALS AND METHODS All patients with SCCOCC treated with radical radiotherapy at an oncology department in Leicester (UK) between 2011 and 2017 were identified. Baseline demographic, clinical data and 2-year treatment outcomes were reported. RESULTS Of the 109 patients included, 40 were South Asian and 59 were non-South Asian. South Asians had significantly poorer 2-year disease-free survival compared with non-South Asians (54.6% versus 73%, P = 0.01). CONCLUSION Our analysis suggests that South Asians with SCCOCC have poorer outcomes despite a younger age and similar disease characteristics. Environmental, social factors and differing biology of disease may be responsible and further research is required to inform targeted interventions.
Collapse
Affiliation(s)
- N Patil
- University Hospitals of Leicester, Leicester, UK.
| | - N Ma
- University Hospitals of Leicester, Leicester, UK
| | - M Mair
- University Hospitals of Leicester, Leicester, UK
| | - J Nazareth
- University Hospitals of Leicester, Leicester, UK
| | - A Sim
- University Hospitals of Leicester, Leicester, UK
| | - C Reynolds
- University Hospitals of Leicester, Leicester, UK
| | - N Freeman
- University Hospitals of Leicester, Leicester, UK
| | - M Chauhan
- University Hospitals of Leicester, Leicester, UK
| | - L Howells
- Institute for Precision Health, University of Leicester, Leicester, UK
| | - D Peel
- Department of Radiation Oncology, Regional Cancer Treatment Services, Midcentral District Health Board, Palmerston North, New Zealand
| | - S Ahmad
- University Hospitals of Leicester, Leicester, UK
| | - T Sridhar
- University Hospitals of Leicester, Leicester, UK
| | - H S Walter
- University Hospitals of Leicester, Leicester, UK; Leicester Cancer Research Centre, University of Leicester, Leicester, UK
| |
Collapse
|
6
|
Lim ZL, Ho PJ, Hartman M, Tan EY, Riza NKBM, Lim EH, Nitar P, Joint Breast Cancer Registry Jbcr, Wong FY, Li J. How Asian Breast Cancer Patients Experience Unequal Incidence of Chemotherapy Side Effects: A Look at Ethnic Disparities in Febrile Neutropenia Rates. Cancers (Basel) 2023; 15:3590. [PMID: 37509253 PMCID: PMC10377556 DOI: 10.3390/cancers15143590] [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: 05/05/2023] [Revised: 06/26/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
The majority of published findings on chemotherapy-induced febrile neutropenia (FN) are restricted to three ethnic groups: Asians, Caucasians, and African Americans. In this two-part study, we examined FN incidence and risk factors in Chinese, Malay, and Indian chemotherapy-treated breast cancer (BC) patients. Hospital records or ICD codes were used to identify patients with FN. In both the Singapore Breast Cancer Cohort (SGBCC) and the Joint Breast Cancer Registry (JBCR), the time of the first FN from the start of chemotherapy was estimated using Cox regression. Multinomial regression was used to evaluate differences in various characteristics across ethnicities. FN was observed in 170 of 1014 patients in SGBCC. The Cox model showed that non-Chinese were at higher risk of developing FN (HRMalay [95% CI]:2.04 [1.44-2.88], p < 0.001; HRIndian:1.88 [1.11-3.18], p = 0.018). In JBCR, FN was observed in 965 of 7449 patients. Univariable Cox models identified ethnicity, a lower baseline absolute neutrophil count, non-luminal A proxy subtypes, and anthracycline-containing regimens as risk factors. Disparities across ethnicities' risk (HRMalay:1.29 [1.07-1.54], p = 0.006; HRIndian:1.50 [1.19-1.88], p < 0.001) remained significant even after further adjustments. Finally, an age-adjusted multinomial model showed that Malays (p = 0.006) and Indians (p = 0.009) were significantly more likely to develop multiple episodes of FN during treatment. Ethnic differences in chemotherapy-induced FN among BC patients exist. Further studies can focus on investigating pharmacogenetic differences across ethnicities.
Collapse
Affiliation(s)
- Zi Lin Lim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Peh Joo Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Mikael Hartman
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Department of Surgery, National University Hospital, Singapore 119054, Singapore
| | - Ern Yu Tan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore 308433, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Singapore 308232, Singapore
| | | | - Elaine Hsuen Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Phyu Nitar
- Department of Cancer Informatics, National Cancer Centre Singapore, Singapore 169610, Singapore
| | | | - Fuh Yong Wong
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jingmei Li
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| |
Collapse
|
7
|
Effects of food and race on the pharmacokinetics of lazertinib in healthy subjects and patients with EGFR mutation-positive advanced non-small cell lung cancer. Lung Cancer 2023; 175:112-120. [PMID: 36495784 DOI: 10.1016/j.lungcan.2022.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVES Lazertinib is a potent, irreversible, brain-penetrant, mutant-selective, and wild type-sparing third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor indicated for advanced non-small cell lung cancer (NSCLC). The study aimed to evaluate the effects of food and race on the pharmacokinetics (PK) of lazertinib from a healthy volunteer trial and PK data from NSCLC patients with EGFR mutation. MATERIALS AND METHODS An open-label, single-dose, two-period, single-sequence crossover study was conducted in healthy subjects with two race groups (non-Asian and Asian). Subjects orally received a single dose of lazertinib 240 mg in fasted and fed state (high-fat meal) in each period separated by a 21-day washout. An open-label, multicenter, phase 1/2 study was conducted in Asian and non-Asian patients with NSCLC. Patients were given oral lazertinib 20-320 mg once daily in fasted state continuously in 21-day cycles. PK parameters were evaluated using non-compartmental analysis. RESULTS A total of 24 healthy subjects (12 non-Asians and 12 Asians) and 52 NSCLC patients (22 non-Asians and 30 Asians) were evaluated. The change in the overall systemic exposure of lazertinib at fed state was less than 15%. Non-Asians showed 58-76% of the systemic exposure than Asians in healthy subjects. In contrast, there were no significant differences in systemic exposure by race both after single and multiple doses among NSCLC patients. CONCLUSION Lazertinib can be taken with or without food considering the comparable systemic exposures related to food. Although effect of race was not consistent across studies, there was no evidence for dose adjustment based on race.
Collapse
|
8
|
Barth A, Hossain M, Perry CR, Gross AS, Ogura H, Shabbir S, Thomas S, Dumont EF, Brimhall DB, Srinivasan M, Swift B. Pharmacokinetic, Safety, and Tolerability Evaluations of Gepotidacin (GSK2140944) in Healthy Japanese Participants. Clin Pharmacol Drug Dev 2023; 12:38-56. [PMID: 36468634 PMCID: PMC10107257 DOI: 10.1002/cpdd.1192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/02/2022] [Indexed: 12/12/2022]
Abstract
Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene antibiotic in late-phase development for uncomplicated urinary tract infection and uncomplicated urogenital gonorrhea. Two clinical studies were conducted to assess the pharmacokinetics (PK) and interethnic comparisons of oral gepotidacin (free-base and to-be-marketed mesylate formulations) administered as single doses ranging from 1500 to 3000 mg in fed and fasted states, and as 2 × 3000-mg doses given 12 hours apart under fed conditions in healthy participants of Japanese ancestry. Dose proportionality was observed in plasma exposures, and comparable area under the concentration-time curve (AUC) and maximum concentration were observed in fed and fasted states. Interethnic comparisons for Japanese versus non-Japanese participant data showed slightly higher plasma maximum concentration (7%-30%) yet similar plasma AUCs; slightly lower urine AUCs (11%-18%) were observed. The slightly higher plasma exposures in healthy Japanese versus White participants in the same study were attributed to lower mean body weights (64 kg versus ≈80 kg). Adverse events were primarily gastrointestinal, and when administered with food, gastrointestinal tolerability was improved. Overall, the gepotidacin PK and safety-risk profiles in healthy Japanese support potential evaluation of the global clinical doses in future studies.
Collapse
Affiliation(s)
- Aline Barth
- GSK, Collegeville, Pennsylvania, USA.,Present affiliation: Global Blood Therapeutics, San Francisco, California, USA
| | - Mohammad Hossain
- GSK, Collegeville, Pennsylvania, USA.,Present affiliation: Servier Pharmaceuticals, Boston, Massachusetts, USA
| | | | | | | | | | | | - Etienne F Dumont
- GSK, Collegeville, Pennsylvania, USA.,Present affiliation: Boston Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
9
|
Low JL, Lim E, Bharwani L, Wong A, Wong K, Ow S, Lim SE, Lee M, Choo J, Lim J, Chan G, Walsh RJ, Muthu V, Ngoi N, Chong W, Tan SH, Lee SC. Real-world outcomes from use of CDK4/6 inhibitors in the management of advanced/metastatic breast cancer in Asia. Ther Adv Med Oncol 2022; 14:17588359221139678. [PMID: 36570409 PMCID: PMC9772978 DOI: 10.1177/17588359221139678] [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: 07/12/2022] [Accepted: 11/01/2022] [Indexed: 12/23/2022] Open
Abstract
Background Oestrogen receptor positive, human epidermal growth factor receptor-2 (HER2) negative breast cancer (BC) is the most frequently diagnosed BC subtype. Combinations of cyclin-dependent kinase 4/6 inhibitors (CDK4/6i) with anti-oestrogen therapy have led to improved survival compared with anti-oestrogen therapy alone for advanced/metastatic BC. The evaluation of CDK4/6i in the real-world facilitates treatment planning, insights into the incidence of drug toxicities, dose modifications including dose delays (DDs) and dose reductions (DRs) and improves prognostic accuracy in subgroups, for example geriatric patients, who are under-represented in clinical trials. Methods This multi-centre study analysed retrospective and prospective data from 456 patients treated with CDK4/6i between January 2015 and December 2020. We examined patient characteristics, variation in prescribing practices, efficacy and toxicity outcomes. Results In all, 456 patients were included in this study. The median age was 59 (range: 24-92). In total, 85 (19%) were ⩾70 years old. In all, 122 (27%) and 119 (26%) of patients were treated in the first-line and second-line settings, respectively. In total, 25 (5%), 31 (7%) and 145 (32%) of patients had brain, peritoneum and liver metastasis, respectively, at the time of CDK4/6i initiation. On univariate analysis, heavily pre-treated patients and those with distant metastases, involving the liver, brain or peritoneum, had significantly shorter progression-free survival (PFS) and 24-month overall survival (OS). Elderly patients (⩾70) had a shorter PFS; OS results were not mature. Majority of patients (n = 362, 80%) initiated treatment with the United States FDA-approved starting dose of CDK4/6i. In all, 330 (72%) had at least one DD and 217 (48%) patients required at least one DR, but these dose modifications were not associated with poorer survival outcomes. Patients age ⩾70 were more likely to require dose modifications leading to a lower treatment dose. The most common reason for DD/DR was neutropenia (60%) and the incidence of febrile neutropenia was only 2%. Conclusions Our study indicates CDK4/6i is effective and safe. Age ⩾ 70, distant metastases to liver, peritoneal or brain were negative prognostic factors. Age ⩾ 70 was associated with significantly increased requirement for dose modification; however, this did not impact survival outcomes. These findings provide reassurance that survival outcomes are not adversely affected in elderly patients when DD/DR is indicated.
Collapse
Affiliation(s)
- Jia Li Low
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Elaine Lim
- Division of Medical Oncology, National Cancer Center Singapore, Singapore
| | - Lavina Bharwani
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | - Andrea Wong
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | | | - Samuel Ow
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Siew Eng Lim
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Matilda Lee
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Joan Choo
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Joline Lim
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Gloria Chan
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Robert John Walsh
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Vaishnavi Muthu
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | - Natalie Ngoi
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | - Wanqin Chong
- Department of Hematology-Oncology, National University Cancer Institute (NCIS), Singapore
| | | | | |
Collapse
|
10
|
The past, present, and future of chemotherapy with a focus on individualization of drug dosing. J Control Release 2022; 352:840-860. [PMID: 36334860 DOI: 10.1016/j.jconrel.2022.10.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022]
Abstract
While there have been rapid advances in developing new and more targeted drugs to treat cancer, much less progress has been made in individualizing dosing. Even though the introduction of immunotherapies such as CAR T-cells and checkpoint inhibitors, as well as personalized therapies that target specific mutations, have transformed clinical treatment of cancers, chemotherapy remains a mainstay in oncology. Chemotherapies are typically dosed on either a body surface area (BSA) or weight basis, which fails to account for pharmacokinetic differences between patients. Drug absorption, distribution, metabolism, and excretion rates can vary between patients, resulting in considerable differences in exposure to the active drugs. These differences result in suboptimal dosing, which can reduce efficacy and increase side-effects. Therapeutic drug monitoring (TDM), genotype guided dosing, and chronomodulation have been developed to address this challenge; however, despite improving clinical outcomes, they are rarely implemented in clinical practice for chemotherapies. Thus, there is a need to develop interventions that allow for individualized drug dosing of chemotherapies, which can help maximize the number of patients that reach the most efficacious level of drug in the blood while mitigating the risks of underdosing or overdosing. In this review, we discuss the history of the development of chemotherapies, their mechanisms of action and how they are dosed. We discuss substantial intraindividual and interindividual variability in chemotherapy pharmacokinetics. We then propose potential engineering solutions that could enable individualized dosing of chemotherapies, such as closed-loop drug delivery systems and bioresponsive biomaterials.
Collapse
|
11
|
Pang A, Jiali L, Ng A, Cheng J, Wang M, Ng YS, Yao Y, Chun M, Ho F, Tey J. Use of the Cancer and Aging Research Group Predictive Model for Chemotherapy-Related Toxic Effects in a Multiethnic, Older Adult Asian Population. JAMA Netw Open 2022; 5:e2237196. [PMID: 36255721 PMCID: PMC9579905 DOI: 10.1001/jamanetworkopen.2022.37196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE The Cancer and Aging Research Group (CARG) prediction model for chemotherapy-related toxic effects has been developed but not yet validated in older Asian adults. In view of differences in drug metabolism and toxic effect reporting in the Asian population, the ability of this tool to guide the cancer treatment decision-making process in older Asian adults needs to be assessed. OBJECTIVE To examine the validity of the CARG predictive model in a multiethnic Asian cohort of older adults. DESIGN, SETTING, AND PARTICIPANTS In this prognostic study, patients of various Asian ethnicities 70 years or older with a solid tumor diagnosis receiving chemotherapy at the National University Cancer Institute, Singapore, were accrued from June 1, 2017, to January 1, 2019. Their risks of chemotherapy-related toxic effects were calculated using the CARG tool. A geriatric assessment was performed, and the treating oncologist (blinded to the CARG scores) was asked to give an estimated likelihood of toxic effects (low, medium, or high). Chemotherapy-related toxic effects were recorded during each clinic visit. Validation of the prediction model was performed by calculating the area under the receiver operating characteristic curve. Multivariate analyses were performed to identify variables in other domains in the geriatric assessment predicting for severe toxic effects. MAIN OUTCOMES AND MEASURES Grade 3 to 5 toxic effects and hospitalization. RESULTS The study included 200 patients (median age, 74 years [range, 70-89 years]; 110 [55.0%] male; 177 [88.5%] Chinese, 17 [8.5%] Malay, 4 [2.0%] Indian, and 2 [1.0%] other ethnicities [according to Singapore's national system of race classification]). A total of 137 patients (68.5%) experienced grade 3 to 5 toxic effects, and 131 (65.5%) required hospitalization. The area under the receiver operating characteristic curve for the CARG chemotoxicity prediction model was 0.74 (95% CI, 0.67-0.82), retaining good discrimination in the study population. CONCLUSIONS AND RELEVANCE This prognostic study conducted in a multiethnic Asian cohort of older adults supports the validity of the CARG predictive model in this population, predicting which older adults are at risk of chemotherapy-related toxic effects.
Collapse
Affiliation(s)
- Angela Pang
- Department of Haematology Oncology, National University Cancer Institute, Singapore
| | - Low Jiali
- Department of Haematology Oncology, National University Cancer Institute, Singapore
| | - Alex Ng
- Yong Loo Lin School of Medicine, National University of Singapore
| | - Joseph Cheng
- Yong Loo Lin School of Medicine, National University of Singapore
| | - Meng Wang
- Department of Medicine, National University Hospital, Singapore
| | - Yean Shin Ng
- Department of Haematology Oncology, National University Cancer Institute, Singapore
| | - Yao Yao
- Department of Pharmacy, National University Hospital, Singapore
| | - Meiling Chun
- Department of Surgery, Ng Teng Fong General Hospital, Singapore
| | - Francis Ho
- Department of Radiation Oncology, National University Cancer Institute, Singapore
| | - Jeremy Tey
- Department of Radiation Oncology, National University Cancer Institute, Singapore
| |
Collapse
|
12
|
Clamp AR, James EC, McNeish IA, Dean A, Kim JW, O'Donnell DM, Gallardo-Rincon D, Blagden S, Brenton J, Perren TJ, Sundar S, Lord R, Dark G, Hall M, Banerjee S, Glasspool RM, Hanna CL, Williams S, Scatchard KM, Nam H, Essapen S, Parkinson C, McAvan L, Swart AM, Popoola B, Schiavone F, Badrock J, Fananapazir F, Cook AD, Parmar M, Kaplan R, Ledermann JA. Weekly dose-dense chemotherapy in first-line epithelial ovarian, fallopian tube, or primary peritoneal cancer treatment (ICON8): overall survival results from an open-label, randomised, controlled, phase 3 trial. Lancet Oncol 2022; 23:919-930. [PMID: 35690073 PMCID: PMC9630160 DOI: 10.1016/s1470-2045(22)00283-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Standard-of-care first-line chemotherapy for epithelial ovarian cancer is carboplatin and paclitaxel administered once every 3 weeks. The JGOG 3016 trial reported significant improvement in progression-free and overall survival with dose-dense weekly paclitaxel and 3-weekly (ie, once every 3 weeks) carboplatin. However, this benefit was not observed in the previously reported progression-free survival results of ICON8. Here, we present the final coprimary outcomes of overall survival and updated progression-free survival analyses of ICON8. METHODS In this open-label, randomised, controlled, phase 3 trial (ICON8), women aged 18 years or older with newly diagnosed stage IC-IV epithelial ovarian, primary peritoneal, or fallopian tube carcinoma (here collectively termed ovarian cancer, as defined by International Federation of Gynecology and Obstetrics [FIGO] 1988 criteria) and an Eastern Cooperative Oncology Group performance status of 0-2 were recruited from 117 hospitals with oncology departments in the UK, Australia and New Zealand, Mexico, South Korea, and Ireland. Patients could enter the trial after immediate primary surgery (IPS) or with planned delayed primary surgery (DPS) during chemotherapy, or could have no planned surgery. Participants were randomly assigned (1:1:1), using the Medical Research Council Clinical Trials Unit at University College London randomisation line with stratification by Gynecologic Cancer Intergroup group, FIGO disease stage, and outcome and timing of surgery, to either 3-weekly carboplatin area under the curve (AUC)5 or AUC6 and 3-weekly paclitaxel 175 mg/m2 (control; group 1), 3-weekly carboplatin AUC5 or AUC6 and weekly paclitaxel 80 mg/m2 (group 2), or weekly carboplatin AUC2 and weekly paclitaxel 80 mg/m2 (group 3), all administered via intravenous infusion for a total of six 21-day cycles. Coprimary outcomes were progression-free survival and overall survival, with comparisons done between group 2 and group 1, and group 3 and group 1, in the intention-to-treat population. Safety was assessed in all patients who started at least one chemotherapy cycle. The trial is registered on ClinicalTrials.gov, NCT01654146, and ISRCTN registry, ISRCTN10356387, and is closed to accrual. FINDINGS Between June 6, 2011, and Nov 28, 2014, 1566 patients were randomly assigned to group 1 (n=522), group 2 (n=523), or group 3 (n=521). The median age was 62 years (IQR 54-68), 1073 (69%) of 1566 patients had high-grade serous carcinoma, 1119 (71%) had stage IIIC-IV disease, and 745 (48%) had IPS. As of data cutoff (March 31, 2020), with a median follow-up of 69 months (IQR 61-75), no significant difference in overall survival was observed in either comparison: median overall survival of 47·4 months (95% CI 43·1-54·8) in group 1, 54·8 months (46·6-61·6) in group 2, and 53·4 months (49·2-59·6) in group 3 (group 2 vs group 1: hazard ratio 0·87 [97·5% CI 0·73-1·05]; group 3 vs group 1: 0·91 [0·76-1·09]). No significant difference was observed for progression-free survival in either comparison and evidence of non-proportional hazards was seen (p=0·037), with restricted mean survival time of 23·9 months (97·5% CI 22·1-25·6) in group 1, 25·3 months (23·6-27·1) in group 2, and 24·8 months (23·0-26·5) in group 3. The most common grade 3-4 adverse events were reduced neutrophil count (78 [15%] of 511 patients in group 1, 183 [36%] of 514 in group 2, and 154 [30%] of 513 in group 3), reduced white blood cell count (22 [4%] in group 1, 80 [16%] in group 2, and 71 [14%] in group 3), and anaemia (26 [5%] in group 1, 66 [13%] in group 2, and 24 [5%] in group 3). No new serious adverse events were reported. Seven treatment-related deaths were reported (two in group 1, four in group 2, and one in group 3). INTERPRETATION In our cohort of predominantly European women with epithelial ovarian cancer, we found that first-line weekly dose-dense chemotherapy did not improve overall or progression-free survival compared with standard 3-weekly chemotherapy and should not be used as part of standard multimodality front-line therapy in this patient group. FUNDING Cancer Research UK, Medical Research Council, Health Research Board in Ireland, Irish Cancer Society, and Cancer Australia.
Collapse
Affiliation(s)
- Andrew R Clamp
- The Christie NHS Foundation Trust and University of Manchester, Manchester, UK
| | - Elizabeth C James
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK.
| | - Iain A McNeish
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Andrew Dean
- Oncology, St John of God Hospital, Subiaco, WA, Australia
| | - Jae-Won Kim
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, South Korea
| | | | | | - Sarah Blagden
- Department of Oncology, Churchill Hospital, University of Oxford, Oxford, UK
| | - James Brenton
- Li Ka Shing Centre, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Tim J Perren
- Leeds Institute of Medical Research, St James' University Hospital, Leeds, UK
| | - Sudha Sundar
- Institute of Cancer and Genomics, University of Birmingham, Birmingham, UK
| | - Rosemary Lord
- Department of Oncology, Clatterbridge Cancer Centre, Wirral, UK
| | - Graham Dark
- Northern Centre for Cancer Care, Freeman Hospital, Newcastle, UK
| | - Marcia Hall
- Department of Medical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - Susana Banerjee
- Gynaecological Unit, The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, UK
| | | | - C Louise Hanna
- Department of Oncology, Velindre Cancer Centre, Cardiff, UK
| | | | - Kate M Scatchard
- North Devon District Hospital, Barnstaple, UK; Exeter Oncology Centre, Royal Devon and Exeter Hospital, Exeter, UK
| | - Helena Nam
- Broomfield Hospital, Chelmsford, UK; Southend University Hospital, Southend, UK
| | - Sharadah Essapen
- St Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, UK
| | | | - Lucy McAvan
- Department of Oncology, University Hospital Coventry and Warwickshire, Coventry, UK
| | - Ann Marie Swart
- Norwich Clinical Trials Unit, Norwich Medical School, University of East Anglia, Norwich, UK
| | - Babasola Popoola
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Francesca Schiavone
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Jonathan Badrock
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Fuad Fananapazir
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Adrian D Cook
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mahesh Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Richard Kaplan
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Jonathan A Ledermann
- Cancer Research UK and UCL Cancer Trials Centre, UCL Cancer Institute and UCL Hospitals, London, UK
| |
Collapse
|
13
|
The Comparative Safety of Epirubicin and Cyclophosphamide versus Docetaxel and Cyclophosphamide in Lymph Node-Negative, HR-Positive, HER2-Negative Breast Cancer (ELEGANT): A Randomized Trial. Cancers (Basel) 2022; 14:cancers14133221. [PMID: 35804991 PMCID: PMC9264845 DOI: 10.3390/cancers14133221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 11/17/2022] Open
Abstract
Background: In adjuvant settings, epirubicin and cyclophosphamide (EC) and docetaxel and cyclophosphamide (TC) are both optional chemotherapy regimens for lymph node-negative, hormone receptor (HR)-positive, human epidermal receptor 2 (HER2)-negative breast cancer patients. Neutropenia is one of the most common adverse events (AEs) of these regimens. The rate of grade 3−4 neutropenia varies in different studies, and direct comparisons of safety profiles between EC and TC are lacking. Method: ELEGANT (NCT02549677) is a prospective, randomized, open-label, noninferior hematological safety trial. Eligible patients with lymph node-negative HR+/HER2-tumors (1:1) were randomly assigned to received four cycles of EC (90/600 mg/m2) or TC (75/600 mg/m2) every three weeks as adjuvant chemotherapy. The primary endpoint was the incidence of grade 3 or 4 neutropenia defined by National Cancer Institute-Common Terminology Criteria for Adverse Events (NCI-CTCAE) version 4.0 on an intention-to-treat basis. Noninferiority was defined as an upper 95% CI less than a noninferiority margin of 15%. Results: In the intention-to-treat population, 140 and 135 patients were randomized into the EC and TC arms, respectively. For the primary endpoint, the rate of grade 3 or 4 neutropenia is 50.71% (95% CI: 42.18%, 59.21%) in the EC arm and 48.15% (95% CI: 39.53%, 56.87%) in the TC arm (95%CI risk difference: −0.100, 0.151), showing the noninferiority of the EC arm. For secondary endpoints, the rate of all-grade anemia is higher in the EC arm (EC 42.86% versus TC 22.96%, p = 0.0007), and more patients suffer from nausea/vomiting, hair loss, and nail changes (p < 0.01) in the EC arm. No statistically different disease-free survival was observed between the two arms (p = 0.13). Conclusion: EC is not inferior to TC in the rate of grade 3 or 4 neutropenia, but more other AEs were observed in the EC group.
Collapse
|
14
|
Makita S, Yamamoto G, Maruyama D, Asano‐Mori Y, Kaji D, Ananthakrishnan R, Ogasawara K, Stepan L, Schusterbauer C, Rettby N, Hasskarl J, Izutsu K. Phase 2 results of lisocabtagene maraleucel in Japanese patients with relapsed/refractory aggressive B-cell non-Hodgkin lymphoma. Cancer Med 2022; 11:4889-4899. [PMID: 35619325 PMCID: PMC9761090 DOI: 10.1002/cam4.4820] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/08/2022] [Accepted: 02/20/2022] [Indexed: 02/03/2023] Open
Abstract
The autologous anti-CD19 chimeric antigen receptor (CAR) T-cell product, lisocabtagene maraleucel (liso-cel), is administered at equal target doses of CD8+ and CD4+ CAR+ T cells. This analysis assessed safety and efficacy of liso-cel in Japanese patients with relapsed or refractory (R/R) aggressive large B-cell lymphoma (LBCL) in Cohort 3 of TRANSCEND WORLD (NCT03484702). Liso-cel (100 × 106 total CAR+ T cells) was administered 2-7 days after lymphodepletion. The primary efficacy endpoint was objective response rate (ORR; Lugano 2014 criteria) assessed by an independent review committee. Fourteen patients were enrolled; 10 received liso-cel infusion (median time to liso-cel availability, 23 days) and were evaluable at data cutoff (median follow-up, 12.5 months). Grade ≥ 3 treatment-emergent adverse events were neutropenia (90%), leukopenia (80%), anemia (70%), and thrombocytopenia (70%). All-grade cytokine release syndrome (CRS) was observed in 50% of patients, though no grade ≥3 CRS events were reported. Grade 1 neurological events occurred in 1 patient but were resolved without any intervention. Prolonged cytopenia (grade ≥ 3 at day 29) was reported for 60% of patients. The ORR was 70%, and complete response rate was 50%. The median duration of response was 9.1 months (95% confidence interval [CI], 2.1-not reached), and overall survival was 14.7 months (95% CI, 1.7-not reached). One patient diagnosed with central nervous system involvement after screening but before liso-cel infusion, responded to liso-cel. Liso-cel demonstrated meaningful efficacy and a manageable safety profile in Japanese patients with R/R LBCL.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Nils Rettby
- Celgene, a Bristol‐Myers Squibb CompanyBoudrySwitzerland
| | - Jens Hasskarl
- Celgene, a Bristol‐Myers Squibb CompanyBoudrySwitzerland
| | | |
Collapse
|
15
|
Chen X, Zhang J, Jiang Q, Yan F. Borrowing historical information to improve phase I clinical trials using meta-analytic-predictive priors. J Biopharm Stat 2022; 32:34-52. [PMID: 35594366 DOI: 10.1080/10543406.2022.2058526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Multiple phase I clinical trials may be performed to determine specific maximum tolerated doses (MTD) for specific races or cancer types. In these situations, borrowing historical information has potential to improve the accuracy of estimating toxicity rate and increase the probability of correctly targeting MTD. To utilize historical information in phase I clinical trials, we proposed using the Meta-Analytic-Predictive (MAP) priors to automatically estimate the heterogeneity between historical trials and give a relatively reasonable amount of borrowed information. We then applied MAP priors in some famous phase I trial designs, such as the continual reassessment method (CRM), Keyboard design and Bayesian optimal interval design (BOIN), to accomplish the process of dose finding. A clinical trial example and extended simulation studies show that our proposed methods have robust and efficient statistical performance, compared with those designs which do not consider borrowing information.
Collapse
Affiliation(s)
- Xin Chen
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jingyi Zhang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qian Jiang
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fangrong Yan
- Department of Biostatistics, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
16
|
Mutation Profiles of Ovarian Seromucinous Borderline Tumors in Japanese Patients. Curr Oncol 2022; 29:3658-3667. [PMID: 35621684 PMCID: PMC9139622 DOI: 10.3390/curroncol29050294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/22/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
Ovarian seromucinous tumors (SMBTs) are relatively rare, and their carcinogenesis is largely unknown. In this study, the molecular features of SMBTs in Japan are assessed. DNA was extracted from microdissected paraffin-embedded sections from 23 SMBT cases. Genetic mutations (KRAS, BRAF, PIK3CA, and ERBB2) were evaluated using Sanger sequencing. Immunohistochemistry for p53, ARID1A, and PTEN was also performed as a surrogate for the loss of functional mutations in these tumor suppressor genes. The prevalence of KRAS, BRAF, PIK3CA, and ERBB2 mutations was 4.3% (1/23), 8.6% (2/23), 8.6% (2/23), and 17.3% (4/23), respectively. Overexpression or loss of p53 expression occurred in 26% (6/23), loss of ARID1A expression in 4.3% (1/23), and none of the cases showed expression of PTEN loss. These findings suggest that KRAS/BRAF/PIK3CA and PTEN mutations are rare carcinogenic events in SMBTs. The high frequency of positive p53 staining and a low frequency of loss of ARID1A staining suggests that SMBT carcinogenesis may be related to the alteration of p53 rather than that of ARID1A. ERBB2 oncogenic mutations may play an important role in the tumorigenesis of Japanese SMBTs.
Collapse
|
17
|
Pramesh CS, Badwe RA, Bhoo-Pathy N, Booth CM, Chinnaswamy G, Dare AJ, de Andrade VP, Hunter DJ, Gopal S, Gospodarowicz M, Gunasekera S, Ilbawi A, Kapambwe S, Kingham P, Kutluk T, Lamichhane N, Mutebi M, Orem J, Parham G, Ranganathan P, Sengar M, Sullivan R, Swaminathan S, Tannock IF, Tomar V, Vanderpuye V, Varghese C, Weiderpass E. Priorities for cancer research in low- and middle-income countries: a global perspective. Nat Med 2022; 28:649-657. [PMID: 35440716 PMCID: PMC9108683 DOI: 10.1038/s41591-022-01738-x] [Citation(s) in RCA: 190] [Impact Index Per Article: 63.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 01/22/2023]
Abstract
Cancer research currently is heavily skewed toward high-income countries (HICs), with little research conducted in, and relevant to, the problems of low- and middle-income countries (LMICs). This regional discordance in cancer knowledge generation and application needs to be rebalanced. Several gaps in the research enterprise of LMICs need to be addressed to promote regionally relevant research, and radical rethinking is needed to address the burning issues in cancer care in these regions. We identified five top priorities in cancer research in LMICs based on current and projected needs: reducing the burden of patients with advanced disease; improving access and affordability, and outcomes of cancer treatment; value-based care and health economics; quality improvement and implementation research; and leveraging technology to improve cancer control. LMICs have an excellent opportunity to address important questions in cancer research that could impact cancer control globally. Success will require collaboration and commitment from governments, policy makers, funding agencies, health care organizations and leaders, researchers and the public.
Collapse
Affiliation(s)
- C S Pramesh
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India.
| | - Rajendra A Badwe
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Nirmala Bhoo-Pathy
- Centre for Epidemiology and Evidence-Based Practice, University of Malaya, Kuala Lumpur, Malaysia
| | - Christopher M Booth
- Departments of Oncology and Public Health Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - Anna J Dare
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | | | - David J Hunter
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Satish Gopal
- Centre for Global Health, National Cancer Institute, Rockville, MD, USA
| | - Mary Gospodarowicz
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | | | | | | | - Peter Kingham
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tezer Kutluk
- Faculty of Medicine and Cancer Institute, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | - Manju Sengar
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | | | | | - Ian F Tannock
- Princess Margaret Cancer Centre and University of Toronto, Toronto, Ontario, Canada
| | | | - Verna Vanderpuye
- National Center for Radiotherapy Oncology and Nuclear Medicine and Korle Bu Teaching Hospital, Accra, Ghana
| | | | | |
Collapse
|
18
|
Karia PS, Tehranifar P, Visvanathan K, Wright JD, Genkinger JM. Cancer-Specific Mortality in Asian American Women Diagnosed with Gynecologic Cancer: A Nationwide Population-Based Analysis. Cancer Epidemiol Biomarkers Prev 2022; 31:578-587. [PMID: 34933960 PMCID: PMC8901555 DOI: 10.1158/1055-9965.epi-21-0829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/07/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cancer is the leading cause of death in Asian Americans (AA), the fastest-growing U.S. population group. Despite heterogeneity in socioeconomic status and health behaviors by ethnicity, few studies have assessed cancer outcomes across AA ethnic groups. We examined differences in gynecologic cancer mortality between AA ethnic groups and non-Hispanic Whites (NHW). METHODS Using the Surveillance, Epidemiology, and End Results database, we identified ovarian (n = 69,113), uterine (n = 157,340), and cervical cancer cases (n = 41,460) diagnosed from 1991-2016. Competing risk regression was used to compare cancer-specific mortality for AAs by ethnicity, using NHW as the reference population. RESULTS In adjusted analyses, AAs had a lower risk of ovarian [HR, 0.90; 95% confidence interval (CI), 0.86-0.94] and cervical cancer death (HR, 0.80; 95% CI, 0.75-0.87) than NHWs, with stronger associations among those ≥50 years at diagnosis [(HRovary, 0.87; 95% CI, 0.82-0.92); (HRcervix, 0.74; 95% CI, 0.67-0.81)]. No overall difference was noted for uterine cancer death (HR, 1.03; 95% CI, 0.97-1.10); however, AAs <50 years at diagnosis had a higher risk of uterine cancer death than NHWs (HR, 1.26; 95% CI, 1.08-1.46). Patterns of cancer mortality were heterogeneous, with Filipino and Chinese women at the highest risk of uterine cancer death and Indian/Pakistani women at the lowest risk of ovarian and cervical cancer death. CONCLUSIONS There are significant differences in gynecologic cancer mortality between AAs and NHWs, with heterogeneity by AA ethnicity. IMPACT Disaggregated analysis of AA is needed to better understand the burden of gynecologic cancer and identify high-risk groups for cancer prevention efforts.
Collapse
Affiliation(s)
- Pritesh S. Karia
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
| | - Parisa Tehranifar
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Jason D. Wright
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY
| | - Jeanine M. Genkinger
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY
| |
Collapse
|
19
|
Joshi A, Patel I, Kapse P, Singh M. Comparative Evaluation of Safety and Efficacy of Alternate Schedule (AS) of Sunitinib in Asian and Non-Asian Patient Population for the Treatment of Metastatic Renal Cell Cancer (mRCC): A Meta-Analysis. KIDNEY CANCER 2022. [DOI: 10.3233/kca-210122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Treatment of metastatic renal cell carcinoma (mRCC) using traditional schedule (TS, 4/2) of Sunitinib is associated with higher adverse effects compared to the alternate schedule (AS, 2/1 upfront or when switched from TS). Objective: This meta-analysis aims to compare the safety, efficacy, and percentage of patients requiring dose reduction or dose interruption between Asian (AP) and non-Asian population (NAP) receiving AS of sunitinib. Methods: Electronic databases (PubMed, EMBASE, Cochrane Library) were searched to identify studies published in the English language between May 2009- May 2019, which included patients (>18 years) with mRCC receiving AS of sunitinib. Data were analyzed using the random effect model and t-test. P < 0.05 was considered statistically significant. Results: Of 1922, 16 studies were included (eight AP, eight NAP). Among all grade AEs, mucositis (RR:0.22; 95% CI:0.12–0.40), cardiotoxicity (RR: 0.52; 95% CI: 0.31–0.88), nausea (RR:0.21; 95% CI: 0.10–0.44), hand-foot syndrome (RR:0.33; 95% CI:0.13–0.83), rash (RR: 0.52; 95% CI: 0.34–0.79), and aspartate transaminase (RR:0.57; 95% CI:0.33–0.98) were more common in AP. Leukopenia (RR:2.57; 95% CI:1.47–4.49), proteinemia (RR:4.45; 95% CI:2.12–9.33), and stomatitis (RR:4.33; 95% CI:2.6–7.23) occurred more commonly in NAP. Further, PFS was significantly longer in NAP, while longer OS was observed in AP (p < 0.001). Dose reduction was significantly higher in AP than NAP (52.08% vs. 40.6%, p = 0.0088). Conclusion: Safety profile of AS of sunitinib was similar with variations in the efficacy, dose reduction between AP and NAP. Sunitinib dose or schedule modification may mitigate AEs and enhance efficacy outcomes in mRCC by extending the treatment duration.
Collapse
Affiliation(s)
- Amit Joshi
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - Ishan Patel
- Medical Affairs, Oncology, Pfizer Emerging Asia, Mumbai, India
| | - Pratiksha Kapse
- Medical Affairs, Oncology, Pfizer Emerging Asia, Mumbai, India
| | - Manmohan Singh
- Regional Oncology Medical Lead, Pfizer Emerging Asia, Mumbai, India
| |
Collapse
|
20
|
Eisenkop SM, Okabe H. Prognostic influence of an early time to chemotherapy following primary cytoreductive surgery for advanced epithelial ovarian cancer. J Gynecol Oncol 2022; 33:e80. [PMID: 36245229 PMCID: PMC9634102 DOI: 10.3802/jgo.2022.33.e80] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 06/15/2022] [Accepted: 08/07/2022] [Indexed: 12/03/2022] Open
Abstract
Objective The current investigation analyzes the prognostic role of the time to chemotherapy (TTC) interval following primary cytoreductive surgery for patients with advanced epithelial ovarian cancer. Methods Characteristics and outcome data for 509 consecutive patients with stage IIIB–IVB ovarian, fallopian tube, and peritoneal cancer who had primary cytoreductive surgery between January 2000 and December 2019 are utilized. A univariate Cox regression determined the association of categorical variables with progression-free survival (PFS) and overall survival (OS). Significant variables (p≤0.05) on univariate analysis were applied to Cox proportional hazard regression. Results The median TTC was 19 days and overall follow-up was 62.2 months. The PFS and OS were 25.5 months and 78.4 months for the study cohort plus 28.4 months and OS 84.5 months for patients rendered grossly disease-free. An early TTC (7–14 vs. 15–21 vs. 22–28 vs. >28 days) was associated with an improved PFS (41.7 vs. 30.6 vs. 18.9 vs. 17.9 months; p<0.001) and OS (132.7 vs. 104.6 vs. 56.5 vs. 48.0 months; p<0.001). The performance status, histology, disease distribution, dimension of residual disease, and categorical plus continuous TTC were predictors of PFS and OS. The use of maintenance therapy was also a predictor of PFS, and the route of chemotherapy administration was a predictor of OS. Conclusions For advanced epithelial ovarian cancer, a TTC of less than 21-days was observed to independently improve the PFS and OS. A 7–14 days TTC trended towards a further extension of the OS. Chemotherapy was administered as early as possible after extensive primary cytoreductive surgery for patients with stage IIIB-IV epithelial ovarian cancer. Early time to chemotherapy improved survival. Complete recovery from surgery before chemotherapy is not essential.
Collapse
Affiliation(s)
| | - Hidefusa Okabe
- Math Question Center, Division of Continuing Education, Harvard University, Cambridge, MA, USA
| |
Collapse
|
21
|
The Recruitment Innovation Center: Developing novel, person-centered strategies for clinical trial recruitment and retention. J Clin Transl Sci 2021; 5:e194. [PMID: 34888064 PMCID: PMC8634298 DOI: 10.1017/cts.2021.841] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 08/11/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Clinical trials continue to face significant challenges in participant recruitment and retention. The Recruitment Innovation Center (RIC), part of the Trial Innovation Network (TIN), has been funded by the National Center for Advancing Translational Sciences of the National Institutes of Health to develop innovative strategies and technologies to enhance participant engagement in all stages of multicenter clinical trials. In collaboration with investigator teams and liaisons at Clinical and Translational Science Award institutions, the RIC is charged with the mission to design, field-test, and refine novel resources in the context of individual clinical trials. These innovations are disseminated via newsletters, publications, a virtual toolbox on the TIN website, and RIC-hosted collaboration webinars. The RIC has designed, implemented, and promised customized recruitment support for 173 studies across many diverse disease areas. This support has incorporated site feasibility assessments, community input sessions, recruitment materials recommendations, social media campaigns, and an array of study-specific suggestions. The RIC’s goal is to evaluate the efficacy of these resources and provide access to all investigating teams, so that more trials can be completed on time, within budget, with diverse participation, and with enough accrual to power statistical analyses and make substantive contributions to the advancement of healthcare.
Collapse
|
22
|
Lee YS, Lee JC, Kim JH, Kim J, Hwang JH. Pharmacoethnicity of FOLFIRINOX versus gemcitabine plus nab-paclitaxel in metastatic pancreatic cancer: a systematic review and meta-analysis. Sci Rep 2021; 11:20152. [PMID: 34635731 PMCID: PMC8505398 DOI: 10.1038/s41598-021-99647-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 09/02/2021] [Indexed: 12/20/2022] Open
Abstract
Treatment outcomes between FOLFIRINOX (5-fluorouracil, leucovorin, irinotecan, and oxaliplatin) and GNP (gemcitabine with albumin-bound paclitaxel) as first-line chemotherapy regimens for metastatic pancreatic cancer (PC) were assessed according to ethnic groups categorized as Western or Asian subgroups. PubMed, EMBASE, and Cochrane library were searched. Thirteen studies were eligible in this meta-analysis. Overall survival was not significantly different between FOLFIRINOX and GNP (HR 1.00, 95% CI 0.83–1.20, P = 0.990). However, the Western subgroup showed a higher survival benefit for FOLFIRINOX over GNP (HR 0.84, 95% CI 0.74–0.95, P = 0.006) whereas the Asian subgroup showed the survival benefit for GNP over FOLFIRINOX (HR 1.29, 95% CI 1.03–1.60, P = 0.030). Progression free survival was not significantly different between the two regimens in the Western subgroup (HR 1.01, 95% CI 0.84–1.20, P = 0.950) and the Asian subgroup (HR 1.13, 95% CI 0.97–1.33, P = 0.110). Occurrence of febrile neutropenia was significantly higher in FOLFIRINOX at both ethnic subgroups; however, that of peripheral neuropathy was significantly higher only in GNP of the Asian subgroup. Therefore, pharmacoethnicity might be a factor worth considering when deciding on a frontline chemotherapeutic regimen although the overall survival was not significantly different between FOLFIRINOX and GNP for metastatic PCs.
Collapse
Affiliation(s)
- Yoon Suk Lee
- Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Republic of Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Jae-Hyeong Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea.
| |
Collapse
|
23
|
Porras AM, Shi Q, Zhou H, Callahan R, Montenegro-Bethancourt G, Solomons N, Brito IL. Geographic differences in gut microbiota composition impact susceptibility to enteric infection. Cell Rep 2021; 36:109457. [PMID: 34320343 PMCID: PMC8333197 DOI: 10.1016/j.celrep.2021.109457] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/26/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022] Open
Abstract
Large-scale studies of human gut microbiomes have revealed broad differences in composition across geographically distinct populations. Yet, studies examining impacts of microbiome composition on various health outcomes typically focus on single populations, posing the question of whether compositional differences between populations translate into differences in susceptibility. Using germ-free mice humanized with microbiome samples from 30 donors representing three countries, we observe robust differences in susceptibility to Citrobacter rodentium, a model for enteropathogenic Escherichia coli infections, according to geographic origin. We do not see similar responses to Listeria monocytogenes infections. We further find that cohousing the most susceptible and most resistant mice confers protection from C. rodentium infection. This work underscores the importance of increasing global participation in microbiome studies related to health outcomes. Diverse cohorts are needed to identify both population-specific responses to specific microbiome interventions and to achieve broader-reaching biological conclusions that generalize across populations.
Collapse
Affiliation(s)
- Ana Maria Porras
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Qiaojuan Shi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Hao Zhou
- Department of Microbiology, Cornell University, Ithaca, NY, USA
| | - Rowan Callahan
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | | | - Noel Solomons
- Center for Studies of Sensory Impairment, Aging and Metabolism (CeSSIAM), Guatemala City, Guatemala
| | - Ilana Lauren Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
24
|
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
|
25
|
Undercutting efforts of precision medicine: roadblocks to minority representation in breast cancer clinical trials. Breast Cancer Res Treat 2021; 187:605-611. [PMID: 34080093 DOI: 10.1007/s10549-021-06264-x] [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: 02/18/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022]
Abstract
Precision (or personalized) medicine holds great promise in the treatment of breast cancer. The success of personalized medicine is contingent upon inclusivity and representation for minority groups in clinical trials. In this article, we focus on the roadblocks for the African American demographic, including the barriers to access and enrollment in breast oncology trials, the prevailing classification of race and ethnicity, and the need to refine monolithic categorization by employing genetic ancestry mapping tools for a more accurate determination of race or ethnicity.
Collapse
|
26
|
Stein JN, Charlot M, Cykert S. Building Toward Antiracist Cancer Research and Practice: The Case of Precision Medicine. JCO Oncol Pract 2021; 17:273-277. [PMID: 33974820 PMCID: PMC8257901 DOI: 10.1200/op.20.01070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jacob N. Stein
- Division of Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC
- Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC
| | - Marjory Charlot
- Division of Oncology, Department of Medicine, University of North Carolina, Chapel Hill, NC
| | - Samuel Cykert
- Division of General Internal Medicine and Clinical Epidemiology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
27
|
Tan VZZ, Chan NM, Ang WL, Mya SN, Chan MY, Chen CK. Cardiotoxicity After Anthracycline Chemotherapy for Childhood Cancer in a Multiethnic Asian Population. Front Pediatr 2021; 9:639603. [PMID: 33614560 PMCID: PMC7888269 DOI: 10.3389/fped.2021.639603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/13/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Anthracyclines are widely used to treat childhood cancers; however, they cause cardiotoxicity. To address the paucity of clinical data from Asian populations, this study investigated the epidemiology of pediatric anthracycline-induced cardiotoxicity, during and after chemotherapy, in a multiethnic Asian population. Procedure: This was a single-center, retrospective analysis of 458 anthracycline-treated pediatric oncology patients at KK Women's and Children's Hospital, a tertiary children's hospital in Singapore from 2005 through 2015. We investigated cardiotoxicity (defined as left ventricular fractional shortening <28% on echocardiography) and its risk factors using univariate logistic regression as well as survival estimates through the Kaplan-Meier method to compare survival distribution between patients with and without cardiotoxicity. Results: Over a follow-up period of almost 4 years, we found that 7% (32/458) of the cohort developed cardiotoxicity, with 37.5% (12/32) of these manifesting as clinical heart failure, whilst the rest were asymptomatic. The cardiotoxic cohort demonstrated a significantly higher mortality rate compared to the non-cardiotoxic group at 46.9 vs. 19.2% (p < 0.001), of whom 3 (9.4%) died from end-stage heart failure. We found that traditional predictors such as female sex, age at diagnosis, and cumulative doxorubicin equivalent dose were not predictors of cardiotoxicity. Conclusion: Our study reaffirms that freedom from symptoms does not ensure normal heart function and suggests that children with abnormal ventricular systolic function have higher mortality risk compared to those with normal systolic function. The findings contribute to improved understanding of the Asian burden to aid development of measures to prevent or reduce the risk of cardiotoxicity.
Collapse
Affiliation(s)
- Varen Zhi Zheng Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nicole Min Chan
- Department of Family Medicine, Tan Tock Seng Hospital, National Healthcare Group, Singapore, Singapore
| | - Wai Lin Ang
- Cardiology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore
| | - Soe Nwe Mya
- Haematology-Oncology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore
| | - Mei Yoke Chan
- Haematology-Oncology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore
| | - Ching Kit Chen
- Cardiology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital, Singapore, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| |
Collapse
|
28
|
Xavier Assad D, Acevedo AC, Cançado Porto Mascarenhas E, Costa Normando AG, Pichon V, Chardin H, Neves Silva Guerra E, Combes A. Using an Untargeted Metabolomics Approach to Identify Salivary Metabolites in Women with Breast Cancer. Metabolites 2020; 10:metabo10120506. [PMID: 33322065 PMCID: PMC7763953 DOI: 10.3390/metabo10120506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/24/2022] Open
Abstract
Metabolic alterations are a hallmark of the malignant transformation in cancer cells, which is characterized by multiple changes in metabolic pathways that are linked to macromolecule synthesis. This study aimed to explore whether salivary metabolites could help discriminate between breast cancer patients and healthy controls. Saliva samples from 23 breast cancer patients and 35 healthy controls were subjected to untargeted metabolomics using liquid chromatography-quadrupole time-of-flight mass spectrometry and a bioinformatics tool (XCMS Online), which revealed 534 compounds, characterized by their retention time in reverse-phase liquid chromatography and by the m/z ratio detected, that were shared by the two groups. Using the METLIN database, 31 compounds that were upregulated in the breast cancer group (p < 0.05) were identified, including seven oligopeptides and six glycerophospholipids (PG14:2, PA32:1, PS28:0, PS40:6, PI31:1, and PI38:7). In addition, pre-treatment and post-treatment saliva samples were analyzed for 10 patients who experienced at least a partial response to their treatment. In these patients, three peptides and PG14:2 were upregulated before but not after treatment. The area under the curve, sensitivity, and specificity for PG14:2 was 0.7329, 65.22%, and 77.14%, respectively. These results provide new information regarding the salivary metabolite profiles of breast cancer patients, which may be useful biomarkers.
Collapse
Affiliation(s)
- Daniele Xavier Assad
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasília Campus Universitário Darcy Ribeiro, Brasília DF 70910-900, Brazil; (D.X.A.); (A.C.A.); (E.C.P.M.); (A.G.C.N.); (E.N.S.G.)
- Medical Oncology Department, Hospital Sírio-Libanês, SGAS 613 Conj. E Bl. B, Brasília DF 70200-730, Brazil
| | - Ana Carolina Acevedo
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasília Campus Universitário Darcy Ribeiro, Brasília DF 70910-900, Brazil; (D.X.A.); (A.C.A.); (E.C.P.M.); (A.G.C.N.); (E.N.S.G.)
| | - Elisa Cançado Porto Mascarenhas
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasília Campus Universitário Darcy Ribeiro, Brasília DF 70910-900, Brazil; (D.X.A.); (A.C.A.); (E.C.P.M.); (A.G.C.N.); (E.N.S.G.)
- Medical Oncology Department, Cettro—Centro de Câncer de Brasília, SMH/N Quadra 02, 12 Andar, Brasilia DF 70710-904, Brazil
| | - Ana Gabriela Costa Normando
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasília Campus Universitário Darcy Ribeiro, Brasília DF 70910-900, Brazil; (D.X.A.); (A.C.A.); (E.C.P.M.); (A.G.C.N.); (E.N.S.G.)
| | - Valérie Pichon
- Department of Analytical, Bioanalytical Sciences and Miniaturization (LSABM), UMR CBI 8231, ESPCI Paris, CNRS, PSL University, 75005 Paris, Ide de France, France; (V.P.); (H.C.)
- Campus UPMC, Sorbonne Université, 75005 Paris, France
| | - Helene Chardin
- Department of Analytical, Bioanalytical Sciences and Miniaturization (LSABM), UMR CBI 8231, ESPCI Paris, CNRS, PSL University, 75005 Paris, Ide de France, France; (V.P.); (H.C.)
- Faculté de Chirurgie Dentaire, Université de Paris, 92120 Montouge, France
| | - Eliete Neves Silva Guerra
- Laboratory of Oral Histopathology, Health Sciences Faculty, University of Brasília Campus Universitário Darcy Ribeiro, Brasília DF 70910-900, Brazil; (D.X.A.); (A.C.A.); (E.C.P.M.); (A.G.C.N.); (E.N.S.G.)
| | - Audrey Combes
- Department of Analytical, Bioanalytical Sciences and Miniaturization (LSABM), UMR CBI 8231, ESPCI Paris, CNRS, PSL University, 75005 Paris, Ide de France, France; (V.P.); (H.C.)
- Correspondence: ; Tel.: +33-1-40-79-46-73
| |
Collapse
|
29
|
Establishment of a bladder cancer cell line expressing both mesenchymal and epithelial lineage-associated markers. Hum Cell 2020; 34:675-687. [PMID: 33165868 DOI: 10.1007/s13577-020-00456-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/29/2020] [Indexed: 10/23/2022]
Abstract
Several experimental models including patient biopsies, animal models, and cell lines have been recommended to study the mechanism of bladder cancer development. After several passages in culture, cell lines lose some original features, and no longer resemble the cells of their original tumor. This makes it necessary to establish various cell lines. In an attempt to establish a new cell line for bladder cancer, JAM-ICR (RRID: CVCL_A9QB) was derived from a 64-year-old man diagnosed with a high-grade tumor. This cell line was characterized in multiple experiments involving morphological studies, immunophenotyping (by immunohistochemistry and flow cytometry), karyotyping, short tandem repeat analysis, colony-forming assays, migration and invasion assays, and chemosensitivity to anti-cancer drugs. JAM-ICR cells are pale with an irregular polygonal shape, and show some similarities to mesenchymal stem cells but with a wider shape and shorter arms. Phenotypic assessment demonstrated the simultaneous expression of mesenchymal-(vimentin, desmin, CD29, CD90, and CD106) and epithelial lineage (pan-cytokeratin) markers, which supports a phenotype similar to epithelial-mesenchymal transition for this cell line. JAM-ICR displayed high metastatic potential and stem-like properties, i.e., self-renewal, colony forming, and the coexpression of TRA-1 with CD44 and CD166. Furthermore, this cell line was significantly more resistant to doxorubicin in comparison to the 5637 cell line. These features make JAM-ICR a new bladder cancer cell line with metastatic potential and stem-like properties, which may be potentially useful as a model to elucidate the molecular and cellular mechanisms of bladder cancer pathogenesis or evaluate new drugs.
Collapse
|
30
|
Matossian MD, Giardina AA, Wright MK, Elliott S, Loch MM, Nguyen K, Zea AH, Lau FH, Moroz K, Riker AI, Jones SD, Martin EC, Bunnell BA, Miele L, Collins-Burow BM, Burow ME. Patient-Derived Xenografts as an Innovative Surrogate Tumor Model for the Investigation of Health Disparities in Triple Negative Breast Cancer. ACTA ACUST UNITED AC 2020; 1:383-392. [PMID: 33786503 PMCID: PMC7784803 DOI: 10.1089/whr.2020.0037] [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] [Accepted: 07/27/2020] [Indexed: 12/24/2022]
Abstract
Despite a decline in overall incidence rates for cancer in the past decade, due in part to impressive advancements in both diagnosis and treatment, breast cancer (BC) remains the leading cause of cancer-related deaths in women. BC alone accounts for ∼30% of all new cancer diagnoses in women worldwide. Triple-negative BC (TNBC), defined as having no expression of the estrogen or progesterone receptors and no amplification of the HER2 receptor, is a subtype of BC that does not benefit from the use of estrogen receptor-targeting or HER2-targeting therapies. Differences in socioeconomic factors and cell intrinsic and extrinsic characteristics have been demonstrated in Black and White TNBC patient tumors. The emergence of patient-derived xenograft (PDX) models as a surrogate, translational, and functional representation of the patient with TNBC has led to the advances in drug discovery and testing of novel targeted approaches and combination therapies. However, current established TNBC PDX models fail to represent the diverse patient population and, most importantly, the specific ethnic patient populations that have higher rates of incidence and mortality. The primary aim of this review is to emphasize the importance of using clinically relevant translatable tumor models that reflect TNBC human tumor biology and heterogeneity in high-risk patient populations. The focus is to highlight the complexity of BC as it specifically relates to the management of TNBC in Black women. We discuss the importance of utilizing PDX models to study the extracellular matrix (ECM), and the distinct differences in ECM composition and biophysical properties in Black and White women. Finally, we demonstrate the crucial importance of PDX models toward novel drug discovery in this patient population.
Collapse
Affiliation(s)
- Margarite D Matossian
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Alexandra A Giardina
- Biospecimen Core Laboratory, Louisiana Cancer Research Center, New Orleans, Louisiana, USA
| | - Maryl K Wright
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Steven Elliott
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Michelle M Loch
- Section of Hematology and Oncology, Department of Medicine, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, Louisiana, USA
| | - Khoa Nguyen
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Arnold H Zea
- Biospecimen Core Laboratory, Louisiana Cancer Research Center, New Orleans, Louisiana, USA.,Department of Genetics and Stanley S. Scott Cancer Center, Louisiana Health Sciences Center, New Orleans, Louisiana, USA
| | - Frank H Lau
- Department of Surgery, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, Louisiana, USA
| | - Krzysztof Moroz
- Biospecimen Core Laboratory, Louisiana Cancer Research Center, New Orleans, Louisiana, USA.,Department of Pathology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Adam I Riker
- Department of Surgery, Louisiana State University Health Sciences Center, School of Medicine, New Orleans, Louisiana, USA.,Department of Surgery, DeCesaris Cancer Institute, Anne Arundel Medical Center, Luminis Health, Annapolis, Maryland, USA
| | - Steven D Jones
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Elizabeth C Martin
- Department of Biological & Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bruce A Bunnell
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Lucio Miele
- Department of Genetics and Stanley S. Scott Cancer Center, Louisiana Health Sciences Center, New Orleans, Louisiana, USA
| | - Bridgette M Collins-Burow
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Matthew E Burow
- Section of Hematology and Oncology, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
31
|
Bass SB, D’Avanzo P, Alhajji M, Ventriglia N, Trainor A, Maurer L, Eisenberg R, Martinez O. Exploring the Engagement of Racial and Ethnic Minorities in HIV Treatment and Vaccine Clinical Trials: A Scoping Review of Literature and Implications for Future Research. AIDS Patient Care STDS 2020; 34:399-416. [PMID: 32931317 PMCID: PMC10722429 DOI: 10.1089/apc.2020.0008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
HIV disproportionately impacts US racial and ethnic minorities but they participate in treatment and vaccine clinical trials at a lower rate than whites. To summarize barriers and facilitators to this participation we conducted a scoping review of the literature guided by the Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Studies published from January 2007 and September 2019 were reviewed. Thirty-one articles were identified from an initial pool of 325 records using three coders. All records were then assessed for barriers and facilitators and summarized. Results indicate that while racial and ethnic minority participation in these trials has increased over the past 10 years, rates still do not proportionately reflect their burden of HIV infection. While many of the barriers mirror those found in other disease clinical trials (e.g., cancer), HIV stigma is a unique and important barrier to participating in HIV clinical trials. Recommendations to improve recruitment and retention of racial and ethnic minorities include training health care providers on the importance of recruiting diverse participants, creating interdisciplinary research teams that better represent who is being recruited, and providing culturally competent trial designs. Despite the knowledge of how to better recruit racial and ethnic minorities, few interventions have been documented using these strategies. Based on the findings of this review, we recommend that future clinical trials engage community stakeholders in all stages of the research process through community-based participatory research approaches and promote culturally and linguistically appropriate recruitment and retention strategies for marginalized populations overly impacted by HIV.
Collapse
Affiliation(s)
- Sarah Bauerle Bass
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, Temple University College of Public Health, Philadelphia, Pennsylvania, USA
| | - Paul D’Avanzo
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, Temple University College of Public Health, Philadelphia, Pennsylvania, USA
| | - Mohammed Alhajji
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, Temple University College of Public Health, Philadelphia, Pennsylvania, USA
| | - Nicole Ventriglia
- Fox Chase Cancer Center, Risk Assessment Program, Philadelphia, Pennsylvania, USA
| | - Aurora Trainor
- Risk Communication Laboratory, Department of Social and Behavioral Sciences, Temple University College of Public Health, Philadelphia, Pennsylvania, USA
| | - Laurie Maurer
- Tennessee Department of Health, HIV/STD/Viral Hepatitis Section, Nashville, Tennessee, USA
| | | | - Omar Martinez
- School of Social Work, Temple University College of Public Health, Philadelphia, Pennsylvania, USA
| |
Collapse
|
32
|
Millet-Boureima C, Selber-Hnatiw S, Gamberi C. Drug discovery and chemical probing in Drosophila. Genome 2020; 64:147-159. [PMID: 32551911 DOI: 10.1139/gen-2020-0037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Flies are increasingly utilized in drug discovery and chemical probing in vivo, which are novel technologies complementary to genetic probing in fundamental biological studies. Excellent genetic conservation, small size, short generation time, and over one hundred years of genetics make Drosophila an attractive model for rapid assay readout and use of analytical amounts of compound, enabling the experimental iterations needed in early drug development at a fraction of time and costs. Here, we describe an effective drug-testing pipeline using adult flies that can be easily implemented to study several disease models and different genotypes to discover novel molecular insight, probes, quality lead compounds, and develop novel prototype drugs.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Susannah Selber-Hnatiw
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
33
|
Tareen SA, Rodriguez J, Bolos D. Retrospective Analysis of HER2+ Breast Cancer Outcomes at a County Hospital: Do Published Outcomes Hold up in the Real World? Cureus 2020; 12:e7937. [PMID: 32499977 PMCID: PMC7266367 DOI: 10.7759/cureus.7937] [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: 11/05/2022] Open
Abstract
INTRODUCTION Landmark trials repeatedly demonstrate that pertuzumab and trastuzumab plus standard chemotherapy have the best outcomes in human epidermal growth factor receptor 2 (HER2) positive breast cancer in the neoadjuvant, adjuvant, and metastatic setting. However, many of these multicenter landmark trials lack diversity and studied largely Caucasian populations. Our goal is to address this under-representation of minorities, and compare pathologic complete response (pCR) rates in our predominantly Hispanic population with HER2 positive breast cancer receiving the same neoadjuvant chemotherapy (NACT) at Olive View-UCLA Medical Center (OVMC) to that of pCR rates observed in the TRYPHAENA trial. METHODS For this retrospective cohort study, we compiled a list of 53 patients aged 18 and older, 52 female and 1 male, with HER2 positive breast cancer identified by fluorescence in situ hybridization treated at OVMC from December 2015 to May 2018. Our population was 57% Hispanic, 13% white, 13% Filipino, 11% Asian, 2% black, and 4% other. The complete list included patients receiving standard neoadjuvant, adjuvant, and metastatic chemotherapy regimens. We analyzed 23 female patients with HER2 positive breast cancer staged I to IIIC, receiving standard NACT (docetaxel, carboplatin, trastuzumab, and pertuzumab). Metastatic HER2 positive breast cancer patients were excluded. The primary outcome studied was pCR rates after receiving NACT. pCR was defined as the absence of invasive cancer cells from tissue samples removed after surgery. Secondary outcomes measured were side effects of chemotherapy. pCR rates and side effects were compared to TRYPHAENA. Data regarding insurance status, breast cancer detection modality, and time to seek medical attention were recorded. RESULTS 50% of our patients who received NACT achieved pCR. Our pCR rates mirrored those observed in the TRYPHAENA trial (51.9%). The most common side effect observed in our population was diarrhea. A higher proportion (37.5%) of our patients had liver function test (LFT) elevation compared to the TRYPHAENA trial (3.9%). Baseline LFTs were normal prior to treatment in 96% of patients. In terms of modality of detection, 70% were self-palpated, 26% were detected through routine mammography, and 4% were found incidentally. Average time from mass discovery to seeking medical attention was 3.4 months. Only 26% had medical insurance at diagnosis. Although not included in our study, 28% of our patients were initially diagnosed with stage IV metastatic disease. Conclusion: Our study found that pCR rates in our primarily Hispanic population compared well to the response rates observed in landmark trials with largely Caucasian populations. Genetic variations in chemo-sensitivity may have a minimal influence on cancer care outcomes in this population.
Collapse
Affiliation(s)
- Serene A Tareen
- Internal Medicine, Olive View - University of California Los Angeles (UCLA) Medical Center, Los Angeles, USA
| | - Joshua Rodriguez
- Hematology & Oncology, Olive View - University of California Los Angeles (UCLA) Medical Center, Los Angeles, USA
| | - David Bolos
- Hematology & Oncology, Olive View - University of California Los Angeles (UCLA) Medical Center, Los Angeles, USA
| |
Collapse
|
34
|
Modi ND, Sorich MJ, Rowland A, McKinnon RA, Koczwara B, Wiese MD, Hopkins AM. Predicting Thrombocytopenia in Patients With Breast Cancer Treated With Ado-trastuzumab Emtansine. Clin Breast Cancer 2019; 20:e220-e228. [PMID: 31892489 DOI: 10.1016/j.clbc.2019.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/06/2019] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Thrombocytopenia is a common and potentially serious adverse event of ado-trastuzumab emtansine (T-DM1) use in patients with advanced breast cancer. However, the risk factors have been minimally explored. Our aim was to develop a clinical prediction model from the clinicopathologic data that would allow for quantification of the personalized risks of thrombocytopenia from T-DM1 usage. MATERIALS AND METHODS Data from 3 clinical trials, EMILIA (a study of trastuzumab emtansine versus capecitabine + lapatinib in participants with HER2 [human epidermal growth factor receptor 2]-positive locally advanced or metastatic breast cancer), TH3RESA (a study of trastuzumab emtansine in comparison with treatment of physician's choice in participants with HER2-positive breast cancer who have received at least two prior regimens of HER2-directed therapy), and MARIANNE [a study of trastuzumab emtansine (T-DM1) plus pertuzumab/pertuzumab placebo versus trastuzumab (Herceptin) plus a taxane in participants with metastatic breast cancer], were pooled. Cox proportional hazard analysis was used to assess the association between the pretreatment clinicopathologic data and grade ≥ 3 thrombocytopenia occurring within the first 365 days of T-DM1 use. A multivariable clinical prediction model was developed using a backward elimination process. RESULTS Of the 1620 participants, 141 (9%) had experienced grade ≥ 3 thrombocytopenia. On univariable analysis, the body mass index, race, presence of brain metastasis, platelet count, white blood cell count, and concomitant corticosteroid use were significantly associated with the occurrence of grade ≥ 3 thrombocytopenia (P < .05). The multivariable prediction model was optimally defined by race (Asian vs. non-Asian) and platelet count (100-220 vs. 220-300 vs. >300 × 109/L). A large discrimination between the prognostic subgroups was observed. The highest risk subgroup (Asian and platelet count of 100-220 cells ×109/L) had a 40% probability of grade ≥ 3 thrombocytopenia within the first 365 days of T-DM1 therapy compared with 2% for the lowest risk subgroup (non-Asian and platelet count > 300 × 109/L). CONCLUSION A clinical prediction model, defined by race and pretreatment platelet count, was able to discriminate subgroups with a significantly different risk of grade ≥ 3 thrombocytopenia after T-DM1 initiation. The model allows for improved interpretation of the personalized risks and risk/benefit ratio of T-DM1.
Collapse
Affiliation(s)
- Natansh D Modi
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ross A McKinnon
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Bogda Koczwara
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; Department of Medical Oncology, Flinders Medical Centre, Adelaide, SA, Australia
| | - Michael D Wiese
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
35
|
Fasinu PS, Rapp GK. Herbal Interaction With Chemotherapeutic Drugs-A Focus on Clinically Significant Findings. Front Oncol 2019; 9:1356. [PMID: 31850232 PMCID: PMC6901834 DOI: 10.3389/fonc.2019.01356] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/18/2019] [Indexed: 11/30/2022] Open
Abstract
One of the most consequential risks associated with the concomitant use of herbal products and chemotherapeutic agents is herb-drug interactions. The risk is higher in patients with chronic conditions taking multiple medications. Herb-drug interaction is particularly undesirable in cancer management because of the precipitous dose-effect relationship and toxicity of chemotherapeutic agents. The most common mechanism of herb-drug interaction is the herbal-mediated inhibition and/or induction of drug-metabolizing enzymes (DME) and/or transport proteins leading to the alteration in the pharmacokinetic disposition of the victim drug. Most mechanistic research has focused on laboratory-based studies, determining the effects of herbal products on DMEs and extrapolating findings to predict clinical relevance; however, not all DME/transporter protein inhibition/induction results in clinical herb-drug interaction. This study reviews relevant literature and identified six herbal products namely echinacea, garlic, ginseng, grapefruit juice, milk thistle, and St John's wort, which have shown interactions with chemotherapeutic agents in humans. This focus on clinically significant herb-drug interaction, should be of interest to the public including practitioners, researchers, and consumers of cancer chemotherapy.
Collapse
Affiliation(s)
- Pius S Fasinu
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Campbell University, Buies Creek, NC, United States
| | - Gloria K Rapp
- Department of Pharmaceutical Sciences, College of Pharmacy & Health Sciences, Campbell University, Buies Creek, NC, United States
| |
Collapse
|
36
|
Vantaku V, Amara CS, Piyarathna DWB, Donepudi SR, Ambati CR, Putluri V, Tang W, Rajapakshe K, Estecio MR, Terris MK, Castro PD, Ittmann MM, Williams SB, Lerner SP, Sreekumar A, Bollag R, Coarfa C, Kornberg MD, Lotan Y, Ambs S, Putluri N. DNA methylation patterns in bladder tumors of African American patients point to distinct alterations in xenobiotic metabolism. Carcinogenesis 2019; 40:1332-1340. [PMID: 31284295 PMCID: PMC6875901 DOI: 10.1093/carcin/bgz128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 05/23/2019] [Accepted: 07/07/2019] [Indexed: 12/31/2022] Open
Abstract
Racial/ethnic disparities have a significant impact on bladder cancer outcomes with African American patients demonstrating inferior survival over European-American patients. We hypothesized that epigenetic difference in methylation of tumor DNA is an underlying cause of this survival health disparity. We analyzed bladder tumors from African American and European-American patients using reduced representation bisulfite sequencing (RRBS) to annotate differentially methylated DNA regions. Liquid chromatography-mass spectrometry (LC-MS/MS) based metabolomics and flux studies were performed to examine metabolic pathways that showed significant association to the discovered DNA methylation patterns. RRBS analysis showed frequent hypermethylated CpG islands in African American patients. Further analysis showed that these hypermethylated CpG islands in patients are commonly located in the promoter regions of xenobiotic enzymes that are involved in bladder cancer progression. On follow-up, LC-MS/MS revealed accumulation of glucuronic acid, S-adenosylhomocysteine, and a decrease in S-adenosylmethionine, corroborating findings from the RRBS and mRNA expression analysis indicating increased glucuronidation and methylation capacities in African American patients. Flux analysis experiments with 13C-labeled glucose in cultured African American bladder cancer cells confirmed these findings. Collectively, our studies revealed robust differences in methylation-related metabolism and expression of enzymes regulating xenobiotic metabolism in African American patients indicate that race/ethnic differences in tumor biology may exist in bladder cancer.
Collapse
Affiliation(s)
- Venkatrao Vantaku
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Chandra Sekhar Amara
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Sri Ramya Donepudi
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Chandrashekar R Ambati
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Vasanta Putluri
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Wei Tang
- Laboratory of Human Carcinogenesis, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Marcos Roberto Estecio
- Center for Cancer Epigenetics, Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Martha K Terris
- Department of Surgery: Urology, Augusta University, Augusta, GA, USA
| | - Patricia D Castro
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
- Human tissue acquisition and pathology shared source, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Michael M Ittmann
- Human tissue acquisition and pathology shared source, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
- Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, TX, USA
| | - Stephen B Williams
- Division of Urology, Department of Surgery, The University of Texas Medical Branch, Galveston, TX, USA
| | - Seth P Lerner
- Scott Department of Urology, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Arun Sreekumar
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Roni Bollag
- Department of Pathology, Augusta University, Augusta, GA, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| | - Michael D Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern, Dallas, TX, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
37
|
Health-Related Quality of Life during Chemoradiation in Locally Advanced Rectal Cancer: Impacts and Ethnic Disparities. Cancers (Basel) 2019; 11:cancers11091263. [PMID: 31466306 PMCID: PMC6770309 DOI: 10.3390/cancers11091263] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/26/2019] [Indexed: 01/04/2023] Open
Abstract
Aims: There is limited data on health-related quality of life (HRQoL) in locally advanced rectal cancer. We assessed HRQoL before, during and after neoadjuvant chemoradiation, correlated this to corresponding clinician-reported adverse events (CR-AEs) and explored disparities between patients of Asian ethnicity versus Caucasians. Correlation between HRQoL and treatment response was also assessed. Methods: A consecutive sample of patients was recruited. HRQoL was assessed with the EORTC QLQ-C30 before chemoradiation, week three of chemoradiation and one-week pre-surgery. Clinical variables including CR-AEs were recorded at these time-points. Patients self-reported socio-demographic variables. Treatment response was assessed by the tumour regression grade. HRQoL data were analysed with multilevel models. Results: Fifty-one patients were recruited. HRQoL completion rates were ≥86%. Cognitive and role functioning worsened significantly during treatment. Emotional, role and social functioning improved significantly at pre-surgery. Fatigue and nausea/vomiting worsened during treatment while fatigue, appetite loss, diarrhoea and financial difficulties improved from treatment to pre-surgery. Almost 30% of the cohort were Asian ethnicity. Differences were found in multiple HRQoL domains between Asians and Caucasians, with Asians faring worse. Significant differences were evident in physical, role and cognitive functioning, and in seven out of the 8 symptom scales. The correlation between patient-reported outcomes and clinician-reported outcomes was weak, with diarrhoea having the strongest correlation (r = 0.58). Vomiting during treatment correlated with poor response, whilst baseline constipation correlated with good response. Conclusion: Chemoradiation for locally advanced rectal cancer affects multiple HRQoL domains. Our findings highlight the importance of psychological aspects of treatment. Significant differences were identified between the Asian and Caucasian populations, with Asians consistently performing worse. Poor correlations between patient and clinician reporting strongly support the inclusion of patient-reported outcomes in clinical studies. HRQoL domains of vomiting and constipation are potential biomarkers of treatment response.
Collapse
|
38
|
Fuh KC, Java JJ, Chan JK, Kapp DS, Monk BJ, Burger RA, Young RC, Alberts DS, McGuire WP, Markman M, Bell J, Ozols RF, Armstrong DK, Aghajanian C, Bookman MA, Mannel RS. Differences in presentation and survival of Asians compared to Caucasians with ovarian cancer: An NRG Oncology/GOG Ancillary study of 7914 patients. Gynecol Oncol 2019; 154:420-425. [PMID: 31229298 DOI: 10.1016/j.ygyno.2019.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE To compare patient/tumor characteristics and outcomes of Asians to Caucasian patients with epithelial ovarian cancer. METHODS Ancillary data were pooled and analyzed from ten prospective randomized front-line Gynecologic Oncology Group clinical trials from 1996 to 2011. Demographic, clinicopathologic features, disease-specific and all-cause survival were analyzed. RESULTS Of 7914 patients, 7641 were Caucasian and 273 Asian. When compared to Caucasians, Asians were younger at trial enrollment, had a better performance status, earlier-stage cancers (17.2% vs. 8.1% with stage I; p < 0.001), and were more likely to be of clear cell (15.8% vs. 6.2%, p < 0.001) and mucinous (3.3% vs. 1.9%, p < 0.001) histology. Asians had an improved 5-year disease-specific survival of 54.1% compared to 46.1% for Caucasians, p = 0.001. In multivariate analysis, the Asian race remained a significant prognostic factor for all-cause survival (HR: 0.84; 95% CI: 0.72-0.99; p = 0.04). Other factors predictive of improved survival included younger age, better performance status, optimal cytoreduction, earlier stage, non-clear cell histology, and lower grade tumors. CONCLUSION Asians enrolled into phase III ovarian cancer clinical trials were younger, with better performance status, earlier-stage of disease, and have a greater number of clear cell and mucinous tumors. After adjusting for these prognostic factors, Asians have a better survival compared to Caucasians.
Collapse
Affiliation(s)
- Katherine C Fuh
- Department of Gynecologic Oncology, Washington University, St Louis, MO 63110, USA.
| | - James J Java
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John K Chan
- Department of Gynecologic Oncology, California Pacific Medical Center/Sutter Cancer Research Consortium/Palo Alto Medical Foundation, San Francisco, CA 94118, USA.
| | - Daniel S Kapp
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA.
| | - Bradley J Monk
- Department of Gynecologic Oncology, Biltmore Cancer Center, Phoenix, AZ 85016, USA.
| | - Robert A Burger
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Robert C Young
- RCY Consulting, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| | | | - William P McGuire
- Virginia Commonwealth University Massey Cancer Center, Richmond, VA 980037, USA.
| | - Maurie Markman
- Eastern Regional Medical Center, Meadowbrook, PA 19046, USA.
| | - Jeffrey Bell
- Ohio Riverside Methodist Hospital, Columbus, OH 43214, USA
| | | | - Deborah K Armstrong
- Department of Medical Oncology, Johns Hopkins Kimmel Cancer Center, Baltimore, MD 21287, USA.
| | | | | | - Robert S Mannel
- Department of Obstetrics and Gynecology, Peggy and Charles Stephenson Cancer Center - University of Oklahoma, Oklahoma City, OK 73104, USA.
| |
Collapse
|
39
|
Poon LHJ, Yu CP, Peng L, Ewig CLY, Zhang H, Li CK, Cheung YT. Clinical ascertainment of health outcomes in Asian survivors of childhood cancer: a systematic review. J Cancer Surviv 2019; 13:374-396. [PMID: 31055708 PMCID: PMC6548762 DOI: 10.1007/s11764-019-00759-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/05/2019] [Indexed: 12/29/2022]
Abstract
PURPOSE Survivorship in children with cancer comes at a cost of developing chronic treatment-related complications. Yet, it is still an under-researched area in Asia, which shares the largest proportion of the global childhood cancer burden given its vast population. This systematic review summarizes existing literature on clinically ascertained health outcomes in Asian survivors of childhood cancer. METHODS A search was conducted on Ovid Medline and EMBASE for studies that focused on survivors of childhood cancer from countries in East and Southeast Asia; adopted post-treatment clinical ascertainment of organ-specific toxicities or/and secondary malignancy. Studies were excluded if health outcomes were assessed during the acute treatment. RESULTS Fifty-nine studies, enrolling a total of 13,442 subjects, were conducted on survivors of leukemia (34%), CNS tumor (14%), and cohorts of survivors with heterogeneous cancer diagnoses (52%). The studies used different medical evaluation methods to assess cardiovascular (15%), metabolic and infertility (32%), and neurological/neurocognitive (20%) outcomes in survivors. The collective findings suggest potential differences in the prevalence of certain late effects (e.g., secondary malignancy and obesity) among Asian and non-Asian populations, which may reflect differences in treatment regimens, practice, genetic variations, or/and socioeconomic disparity. CONCLUSIONS We recommend developing collaborative initiatives to build a regional repository of systematically assessed health outcomes and biospecimens to investigate treatment, social-environmental and genetic predictors, and interventions for late effects in this population. IMPLICATIONS FOR CANCER SURVIVORS The existing types of chronic health problems identified in this review suggest the need for active screening, better access to survivorship care, and promotion of protective health behavior in Asia.
Collapse
Affiliation(s)
- Long Hin Jonathan Poon
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, 8th Floor, Lo Kwee-Seong Integrated Biomedical Sciences Building, Shatin, N.T, Hong Kong
| | - Chun-Pong Yu
- Li Ping Medical Library, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Liwen Peng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, 8th Floor, Lo Kwee-Seong Integrated Biomedical Sciences Building, Shatin, N.T, Hong Kong
| | - Celeste Lom-Ying Ewig
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, 8th Floor, Lo Kwee-Seong Integrated Biomedical Sciences Building, Shatin, N.T, Hong Kong
| | - Hui Zhang
- Department of Pediatric Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Chi-Kong Li
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- Paediatric Haematology & Oncology, Hong Kong Children's Hospital, Hong Kong, Hong Kong
| | - Yin Ting Cheung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, 8th Floor, Lo Kwee-Seong Integrated Biomedical Sciences Building, Shatin, N.T, Hong Kong.
| |
Collapse
|
40
|
Ghaderi F, Mehdipour F, Hosseini A, Talei A, Ghaderi A. Establishment and Characterization of a New Triple Negative Breast Cancer Cell Line from an Iranian Breast Cancer Tissue. Asian Pac J Cancer Prev 2019; 20:1683-1689. [PMID: 31244288 PMCID: PMC7021626 DOI: 10.31557/apjcp.2019.20.6.1683] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 05/24/2019] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the most common malignancy and the leading cause of cancer-related death among women worldwide. The underlying mechanisms for breast cancer development, especially in young women, are not completely understood. Although there are several experimental models to understand the biology of breast cancer such as immortalized cell lines, many of these cell lines have been in culture for decades and most of them have been derived from Caucasians or African-Americans. So, it is required to establish a new cell line derived from primary tumors and Asian women. In this study Pari-Institute for Cancer Research (Pari-ICR) was derived from the primary breast tumor of a 36-years old patient with invasive ductal carcinoma. We characterized the cell line by examining morphology, expression of different markers, and functional profile. Immunocytochemistry showed that this cell line does not express estrogen and progesterone receptors as well as human epidermal growth factor receptor 2 (HER2). Pari-ICR cell line expresses high levels of Vimentin, Ezrin, and S100 but does not express EpCAM, Cytokeratin19, Pan-cytokeratin, Nestin, and Desmin. Its doubling time of Pari-ICR was about 22h and was able to grow as colonies in soft agar. It displayed a higher ability of migration and invasion in comparison with MCF-7 cell line. This breast cancer cell line can serve as a model for understanding the molecular mechanisms of breast carcinogenesis. Moreover, it can be used as an appropriate resource to find novel biomarkers or assess new drugs.
Collapse
Affiliation(s)
- Farzaneh Ghaderi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Mehdipour
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Hosseini
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abdolrasoul Talei
- Department of Surgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
41
|
Nazha B, Mishra M, Pentz R, Owonikoko TK. Enrollment of Racial Minorities in Clinical Trials: Old Problem Assumes New Urgency in the Age of Immunotherapy. Am Soc Clin Oncol Educ Book 2019; 39:3-10. [PMID: 31099618 DOI: 10.1200/edbk_100021] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Minority U.S. populations are underrepresented in cancer clinical trials. This review appraises the impact of the disparity in clinical trial participation by minority patients in the current era of cancer immunotherapy. Enrollment on pivotal trials leading to U.S. regulatory approval of immune checkpoint inhibitors showed poor representation of minority ethnic groups. Specifically, we found that black patients constitute less than 4% of all patients enrolled across multiple trials that supported the approval of immune checkpoint inhibitors for the treatment of lung cancer. Similar underrepresentation was observed for trials conducted in renal cell carcinoma and other tumor types. Since efficacy of immunotherapy is only observed in a subset of patients, the use of predictive biomarkers to identify responders along with new strategies to expand the benefit to a larger subset of patients are current areas of active investigation. The inadequate representation of minority patients on immunotherapy clinical trials could perpetuate outcome disparity because the unique biology of the host and the tumors from this subpopulation is not accounted for as new treatment algorithms to guide optimal use of immunotherapy are developed for use in the real world.
Collapse
Affiliation(s)
- Bassel Nazha
- 1 Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA
| | - Manoj Mishra
- 2 Department of Biology, Alabama State University, Montgomery, AL
| | - Rebecca Pentz
- 1 Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA
| | - Taofeek K Owonikoko
- 1 Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
42
|
Lee ATJ, Jones RL, Huang PH. Pazopanib in advanced soft tissue sarcomas. Signal Transduct Target Ther 2019; 4:16. [PMID: 31123606 PMCID: PMC6522548 DOI: 10.1038/s41392-019-0049-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/08/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Pazopanib is the first and only tyrosine kinase inhibitor currently approved for the treatment of multiple histological subtypes of soft tissue sarcoma (STS). Initially developed as a small molecule inhibitor of vascular endothelial growth factor receptors, preclinical work indicates that pazopanib exerts an anticancer effect through the inhibition of both angiogenic and oncogenic signaling pathways. Following the establishment of optimal dosing and safety profiles in early phase studies and approval for the treatment of advanced renal cell carcinoma, pazopanib was investigated in STS. A landmark phase III randomized study demonstrated improved progression-free survival with pazopanib compared to that with placebo in pretreated patients with STS of various subtypes. The efficacy of pazopanib in specific STS subtypes has been further described in real-world-based case series in both mixed and subtype-specific STS cohorts. At present, there are no clinically validated predictive biomarkers for use in selecting patients with advanced STS for pazopanib therapy, limiting the clinical effectiveness and cost-effectiveness of the drug. In this review, we summarize the preclinical and clinical data for pazopanib, outline the evidence base for its effect in STS and explore reported studies that have investigated putative biomarkers.
Collapse
Affiliation(s)
- Alex T. J. Lee
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
43
|
Lim H, Kim SY, Lee E, Lee S, Oh S, Jung J, Kim KS, Moon A. Sex-Dependent Adverse Drug Reactions to 5-Fluorouracil in Colorectal Cancer. Biol Pharm Bull 2019; 42:594-600. [DOI: 10.1248/bpb.b18-00707] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hyesol Lim
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University
| | - Sun Young Kim
- Department of Chemistry, College of Natural Sciences, Duksung Women’s University
| | - Eunhye Lee
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University
| | - Seungeun Lee
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University
| | - Sungryong Oh
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University
| | - Joohee Jung
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University
| | - Kwi Suk Kim
- Department of Pharmacy, Seoul National University Hospital
| | - Aree Moon
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women’s University
| |
Collapse
|
44
|
Richardson PG, San Miguel JF, Moreau P, Hajek R, Dimopoulos MA, Laubach JP, Palumbo A, Luptakova K, Romanus D, Skacel T, Kumar SK, Anderson KC. Interpreting clinical trial data in multiple myeloma: translating findings to the real-world setting. Blood Cancer J 2018; 8:109. [PMID: 30413684 PMCID: PMC6226527 DOI: 10.1038/s41408-018-0141-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/26/2018] [Accepted: 10/09/2018] [Indexed: 01/04/2023] Open
Abstract
Substantial improvements in survival have been seen in multiple myeloma (MM) over recent years, associated with the introduction and widespread use of multiple novel agents and regimens, as well as the emerging treatment paradigm of continuous or long-term therapy. However, these therapies and approaches may have limitations in the community setting, associated with toxicity burden, patient burden, and other factors including cost. Consequently, despite improvements in efficacy in the rigorously controlled clinical trials setting, the same results are not always achieved in real-world practice. Furthermore, the large number of different treatment options and regimens under investigation in various MM settings precludes the feasibility of obtaining head-to-head clinical trial data, and there is a temptation to use cross-trial comparisons to evaluate data across regimens. However, multiple aspects, including patient-related, disease-related, and treatment-related factors, can influence clinical trial outcomes and lead to differences between studies that may confound direct comparisons between data. In this review, we explore the various factors requiring attention when evaluating clinical trial data across available agents/regimens, as well as other considerations that may impact the translation of these findings into everyday MM management. We also investigate discrepancies between clinical trial efficacy and real-world effectiveness through a literature review of non-clinical trial data in relapsed/refractory MM on novel agent-based regimens and evaluate these data in the context of phase 3 trial results for recently approved and commonly used regimens. We thereby demonstrate the complexity of interpreting data across clinical studies in MM, as well as between clinical studies and routine-care analyses, with the aim to help clinicians consider all the necessary issues when tailoring individual patients' treatment approaches.
Collapse
Affiliation(s)
| | - Jesus F San Miguel
- Centro Investigación Medica Aplicada (CIMA), IDISNA; CIBERONC, Clinica Universidad de Navarra, Pamplona, Spain
| | - Philippe Moreau
- Department of Hematology, University Hospital Hôtel Dieu, Nantes, France
| | - Roman Hajek
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Meletios A Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Jacob P Laubach
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Antonio Palumbo
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Katarina Luptakova
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Dorothy Romanus
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Tomas Skacel
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
45
|
Zeng X, Luo T, Li J, Li G, Zhou D, Liu T, Zou X, Pandey A, Luo Z. Transcriptomics-based identification and characterization of 11 CYP450 genes of Panax ginseng responsive to MeJA. Acta Biochim Biophys Sin (Shanghai) 2018; 50:1094-1103. [PMID: 30321253 DOI: 10.1093/abbs/gmy120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Indexed: 11/14/2022] Open
Abstract
Cytochromes P450 (CYP450s), a superfamily of mono-oxygenases, are essential to generate highly functionalized secondary metabolites in plants and contribute to the diversification of specialized triterpenoid biosynthesis in eudicots. However, screening and identifying the exact CYP450 genes in ginsenoside biosynthesis is extremely challenging due to existence of large quantities of members in CYP450 superfamily. Therefore, to screen the CYP450 genes involved in ginsenoside biosynthesis, transcriptome dataset of Panax ginseng was created in our previous work using the technique of the next-generation sequencing. On the basis of bioinformatics analysis, 16 putative CYP450 genes with significant differential expression were screened from the dataset and submitted to GenBank, in which 11 of them have been cloned. Methyl jasmonate (MeJA) was used as an elicitor to analyze the expression profiles of candidate CYP450 genes by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). The results of qRT-PCR analysis revealed that the expression of some CYP450 genes were strongly induced by MeJA and showed different transcription levels at different treatment time points. Homology analysis indicated that each putative CYP450 protein of P. ginseng has a conserved domain consisting of E-E-R-F-P-R-G. The CYP450 genes were screened and cloned here to enrich the resources of CYP450 genes, and the results of bioinformatics analysis provided a foundation to further identify the function of CYP450s involved in ginsenoside biosynthesis. Furthermore, this study facilitated the construction of microbial cell factories for increasing the production of ginsenosides by means of metabolic engineering.
Collapse
Affiliation(s)
- Xu Zeng
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Tiao Luo
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Jijia Li
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Gui Li
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Donghua Zhou
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Tuo Liu
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Xian Zou
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Aparna Pandey
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| | - Zhiyong Luo
- Molecular Biology Research Center, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
46
|
Cowan RA, Tseng J, Ali N, Dearie H, Murthy V, Gennarelli RL, Iasonos A, Abu-Rustum NR, Chi DS, Long Roche KC, Brown CL. Exploring the impact of income and race on survival for women with advanced ovarian cancer undergoing primary debulking surgery at a high-volume center. Gynecol Oncol 2018; 149:43-48. [PMID: 29605049 DOI: 10.1016/j.ygyno.2017.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/03/2017] [Accepted: 11/07/2017] [Indexed: 10/17/2022]
Abstract
OBJECTIVE To evaluate patients with advanced ovarian cancer (OC) undergoing primary debulking surgery (PDS) at a high-volume center (HVC), to determine whether socio-demographic disparities in PDS outcome and overall survival (OS) were present. METHODS All patients with stages IIIB-IV high-grade OC undergoing PDS at our institution from 1/2001-12/2013 were identified. Patients self-identified race/ethnicity as non-Hispanic White (NHW), non-Hispanic Black (NHB), Asian (A), or Hispanic (H). Income level for the entire cohort was estimated using the census-reported income level for each patient's zip code as a proxy for SES. Main outcome measures were PDS outcome and median OS. Cox proportional hazards model was used to examine differences in OS by racial/ethnic and income category, controlling for selected clinical factors. RESULTS 963 patients were identified for analysis: 855 NHW; 43 A, 34H, 28 NHB, and 3 unknown. PDS outcome was not significantly different among NHB and H as compared to NHW. Compared to NHW, Asians were more likely to have >1cm residual (AOR 2.32, 95%CI 1.1-4.9, p=0.03). Median income for the entire cohort was $85,814 (range $10,926-$231,667). After adjusting for significant prognostic factors, there were no significant differences in PDS outcome between income groups (p=0.7281). Median OS was 55.1mos (95%CI 51.8-58.5) with no significant differences in OS between the income (p=0.628) or racial/ethnic (p=0.615) groups. CONCLUSION Statistically significant socio-demographic disparities in PDS and survival outcomes were not observed among women with advanced OC treated at this HVC. Increased efforts are needed to centralize care to and increase the diversity of pts treated at HVCs.
Collapse
Affiliation(s)
- Renee A Cowan
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Jill Tseng
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Narisha Ali
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helen Dearie
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vijayashree Murthy
- Department of Surgery, Saint Barnabas Medical Center, Livingston, NJ, USA
| | - Renee L Gennarelli
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, USA
| | - Alexia Iasonos
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, USA
| | - Nadeem R Abu-Rustum
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Dennis S Chi
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Kara C Long Roche
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA
| | - Carol L Brown
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
47
|
Yen CJ, Muro K, Kim TW, Kudo M, Shih JY, Lee KW, Chao Y, Kim SW, Yamazaki K, Sohn J, Cheng R, Zhang Y, Binder P, Mi G, Orlando M, Chung HC. Ramucirumab Safety in East Asian Patients: A Meta-Analysis of Six Global, Randomized, Double-Blind, Placebo-Controlled, Phase III Clinical Trials. J Glob Oncol 2018; 4:1-12. [PMID: 30085888 PMCID: PMC6223521 DOI: 10.1200/jgo.17.00227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose Several ramucirumab trials have reported a higher incidence of selected adverse events (AEs) in East Asian (EA) patients with cancer versus non-EA patients. A meta-analysis was conducted across six completed phase III trials to establish the safety parameters of ramucirumab in EA compared with non-EA patients. Materials and Methods Six global, randomized, double-blind, placebo-controlled, phase III registration trials investigating ramucirumab were assessed. Relative risks (RRs) and 95% CIs were calculated for selected all-grade and grade ≥ 3 AEs using fixed-effects and mixed-effects models. Ratio of RR and number needed to harm were calculated for AEs (all grade and grade ≥ 3) between EA and non-EA patients. Results Of 4,996 randomly assigned patients receiving ramucirumab or placebo, 802 (16.1%) were EA (ramucirumab, n = 411; placebo, n = 391) and 4,194 were non-EA (ramucirumab, n = 2,337; placebo, n = 1,857). Patient baseline characteristics were generally balanced between treatment arms in EA and non-EA patients, excluding sex and body weight. Grade ≥ 3 AEs possibly associated with ramucirumab, which were increased in EA versus non-EA patients, included neutropenia (42.1% v 25.5%, respectively) and proteinuria (3.9% v 0.6%, respectively). There was an increase in the RR of several grade ≥ 3 AEs, including hypertension and proteinuria, in ramucirumab-treated EA and non-EA patients compared with placebo. The ratio of RR revealed no significant differences between EA and non-EA patients for all-grade and grade ≥ 3 AEs. Conclusion Despite the enhanced propensity of selected AEs in EA patients relative to non-EA patients, there were no substantial differences in the RR for AEs possibly associated with ramucirumab in these phase III trials.
Collapse
Affiliation(s)
- Chia-Jui Yen
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Kei Muro
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Tae-Won Kim
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Masatoshi Kudo
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Jin-Yuan Shih
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Keun-Wook Lee
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Yee Chao
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Sang-We Kim
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Kentaro Yamazaki
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - JooHyuk Sohn
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Rebecca Cheng
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Yawei Zhang
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Polina Binder
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Gu Mi
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Mauro Orlando
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| | - Hyun Cheol Chung
- Chia-Jui Yen, National Cheng Kung University Hospital, Tainan; Jin-Yuan Shih, National Taiwan University Hospital; Yee Chao, National Yang-Ming University and Taipei Veterans General Hospital; Rebecca Cheng, Eli Lilly and Company, Taipei, Taiwan; Kei Muro, Aichi Cancer Center Hospital, Nagoya; Masatoshi Kudo, Kindai University School of Medicine, Osaka-Sayama City, Osaka; Kentaro Yamazaki, Shizuoka Cancer Center, Shizuoka, Japan; Tae-Won Kim and Sang-We Kim, Asan Medical Center; JooHyuk Sohn and Hyun Cheol Chung, Yonsei University College of Medicine, Seoul; Keun-Wook Lee, Seoul National University College of Medicine, Seongnam, South Korea; Yawei Zhang and Polina Binder, Eli Lilly and Company, Bridgewater, NJ; Gu Mi, Eli Lilly and Company, Indianapolis, IN, USA; and Mauro Orlando, Eli Lilly and Company, Buenos Aires, Argentina
| |
Collapse
|
48
|
Safety of pazopanib and sunitinib in treatment-naive patients with metastatic renal cell carcinoma: Asian versus non-Asian subgroup analysis of the COMPARZ trial. J Hematol Oncol 2018; 11:69. [PMID: 29788981 PMCID: PMC5964681 DOI: 10.1186/s13045-018-0617-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/06/2018] [Indexed: 01/01/2023] Open
Abstract
Background The international, phase 3 COMPARZ study demonstrated that pazopanib and sunitinib have comparable efficacy as first-line therapy in patients with advanced renal cell carcinoma, but that safety and quality-of-life profiles favor pazopanib. Our report analyzed pazopanib and sunitinib safety in Asian and non-Asian subpopulations. Methods Patients were randomized 1:1 to receive pazopanib 800 mg once daily (continuous dosing) or sunitinib 50 mg once daily in 6-week cycles (4 weeks on, 2 weeks off). Results Safety population was composed of 363 Asian patients and 703 non-Asian patients. Asian patients had similar duration of exposure to either drug compared with non-Asian patients, although Asian patients had a higher frequency of dose modifications. Overall, hematologic toxicities, cytopenias, increased AST/ALT, and palmar-plantar erythrodysesthesia (PPE) were more prevalent in Asian patients, whereas gastrointestinal toxicities were more prevalent in non-Asian patients. Among Asian patients, hematologic adverse events and most non-hematologic AEs were more common in sunitinib-treated versus pazopanib-treated patients. Among Asian patients, the most common grade 3/4 AEs with pazopanib were hypertension (grade 3, 22%) and alanine aminotransferase increased (grade 3, 12%; grade 4, 1%); the most common grade 3/4 AEs with sunitinib were thrombocytopenia/platelet count decreased (grade 3, 36%; grade 4, 10%), neutropenia/neutrophil count decreased (grade 3, 24%; grade 4, 3%) hypertension (grade 3, 20%), and PPE (grade 3, 15%). Conclusions A distinct pattern and severity of adverse events was observed in Asians when compared with non-Asians with both pazopanib and sunitinib. However, the two drugs were well tolerated in both subpopulations. Trial registration ClinicalTrials.gov, NCT00720941, Registered July 22, 2008 ClinicalTrials.gov, NCT01147822, Registered June 22, 2010 Electronic supplementary material The online version of this article (10.1186/s13045-018-0617-1) contains supplementary material, which is available to authorized users.
Collapse
|
49
|
Rossi A, Novello S. The Tradition of the Rising Sun: When Geography Counts. J Thorac Oncol 2018; 13:598-600. [PMID: 29703535 DOI: 10.1016/j.jtho.2018.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 03/15/2018] [Indexed: 11/28/2022]
Affiliation(s)
- Antonio Rossi
- Division of Medical Oncology, Scientific Institute for Research and Health Care Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Silvia Novello
- University of Turin, Department of Oncology, San Luigi Hospital, Orbassano, Turin, Italy.
| |
Collapse
|
50
|
Aghili M, Sotoudeh S, Ghalehtaki R, Babaei M, Farazmand B, Fazeli MS, Keshvari A, Haddad P, Farhan F. Preoperative short course radiotherapy with concurrent and consolidation chemotherapies followed by delayed surgery in locally advanced rectal cancer: preliminary results. Radiat Oncol J 2018; 36:17-24. [PMID: 29621870 PMCID: PMC5903358 DOI: 10.3857/roj.2017.00185] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 10/15/2017] [Accepted: 10/30/2017] [Indexed: 12/22/2022] Open
Abstract
PURPOSE This study aimed to assess complications and outcomes of a new approach, that is, combining short course radiotherapy (SRT), concurrent and consolidative chemotherapies, and delayed surgery. MATERIALS AND METHODS In this single arm phase II prospective clinical trial, patients with T3-4 or N+ M0 rectal adenocarcinoma were enrolled. Patients who received induction chemotherapy or previous pelvic radiotherapy were excluded. Study protocol consisted of three-dimensional conformal SRT (25 Gy in 5 fractions in 1 week) with concurrent and consolidation chemotherapies including capecitabine and oxaliplatin. Total mesorectal excision was done at least 8 weeks after the last fraction of radiotherapy. Primary outcome was complete pathologic response and secondary outcomes were treatment related complications. RESULTS Thirty-three patients completed the planned preoperative chemoradiation and 26 of them underwent surgery (24 low anterior resection and 2 abdominoperineal resection). Acute proctitis grades 2 and 3 were seen in 11 (33.3%) and 7 (21.2%) patients, respectively. There were no grades 3 and 4 subacute hematologic and non-hematologic (genitourinary and peripheral neuropathy) toxicities and perioperative morbidities such as anastomose leakage. Grade 2 or higher late toxicities were observed among 29.6% of the patients. Complete pathologic response was achieved in 8 (30.8%) patients who underwent surgery. The 3-year overall survival and local control rates were 65% and 94%, respectively. CONCLUSION This study showed that SRT combined with concurrent and consolidation chemotherapies followed by delayed surgery is not only feasible and tolerable without significant toxicity but also, associated with promising complete pathologic response rates.
Collapse
Affiliation(s)
- Mahdi Aghili
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sarvazad Sotoudeh
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Ghalehtaki
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Babaei
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Borna Farazmand
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Sadegh Fazeli
- Colorectal Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Keshvari
- Colorectal Research Center, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Peiman Haddad
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Farhan
- Radiation Oncology Research Center, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|