1
|
Dhir A, Hayashi M, Bodlak A, Oesterheld J, Loeb DM, Mascarenhas L, Isakoff MS, Sandler ES, Borinstein SC, Trucco M, Lagmay JP, Setty BA, Pratilas CA, Caywood E, Metts J, Yin H, Fridley B, Yin J, Laborde J, Reed DR, Adams DL, Wagner LM. Phase II Trial of Gemcitabine and Nab-Paclitaxel for Recurrent Osteosarcoma with Serial Monitoring Using Liquid Biopsy: A Report from the National Pediatric Cancer Foundation. Clin Cancer Res 2024; 30:5314-5322. [PMID: 39360936 DOI: 10.1158/1078-0432.ccr-24-1339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/02/2024] [Accepted: 10/01/2024] [Indexed: 12/07/2024]
Abstract
PURPOSE The combination of gemcitabine and docetaxel is often used to treat patients with recurrent osteosarcoma. Nab-paclitaxel has preclinical activity against osteosarcoma and is potentially less myelosuppressive than docetaxel. We conducted a prospective multi-institutional phase II trial combining gemcitabine and nab-paclitaxel for patients aged 12 to 30 years with recurrent osteosarcoma and measurable disease. PATIENTS AND METHODS A Simon's two-stage design was used to test a 4-month progression-free survival (PFS-4) of 10% vs. 35%. Patients received nab-paclitaxel 125 mg/m2 and gemcitabine 1,000 mg/m2 weekly × 3 in 4-week cycles. Immunohistochemical analysis of archival tissue and serial assessment of circulating tumor cells (CTC) and circulating tumor DNA (ctDNA) using ultralow passage whole-genome sequencing were performed to identify potential biomarkers of response. RESULTS Eighteen patients received 56 total cycles (median 2, range 1-12). Two patients (11%) experienced confirmed partial response and six (33%) received >2 cycles. The PFS-4 was 28% (95% confidence interval, 13%-59%). Six patients required dose reductions and three patients were removed due to toxicities. All 18 patients had detectable CTCs and 10 had ctDNA identified. All eight patients with MYC amplification at study entry experienced disease progression. CONCLUSIONS Gemcitabine and nab-paclitaxel demonstrated similar clinical activity and toxicity compared to previous retrospective reports utilizing gemcitabine and docetaxel in patients with recurrent osteosarcoma. Serial analysis of CTC and ctDNA was feasible in this prospective multi-institution study and provides preliminary data on the use of these assays in patients with relapsed disease.
Collapse
Affiliation(s)
- Aditi Dhir
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida
| | - Masanori Hayashi
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Center for Cancer and Blood Disorders Children's Hospital Colorado, Aurora, Colorado
| | - Avery Bodlak
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Center for Cancer and Blood Disorders Children's Hospital Colorado, Aurora, Colorado
| | - Javier Oesterheld
- Cancer and Blood Disorders Program, Atrium Health Levine Children's Hospital, Charlotte, North Carolina
| | - David M Loeb
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Leo Mascarenhas
- Cedar-Sinai Medical Center, Guerin Children's and Cedar-Sinai Cancer, Los Angeles, California
| | - Michael S Isakoff
- Division of Hematology/Oncology, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Eric S Sandler
- Division of Hematology/Oncology, Nemours Children's Health, Jacksonville, Florida
| | - Scott C Borinstein
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Matteo Trucco
- Department of Pediatric Hematology-Oncology and BMT, Cleveland Clinic Children's, Cleveland, Ohio
| | - Joanne P Lagmay
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Shands Childrens Hospital, University of Florida, Gainesville, Florida
| | - Bhuvana A Setty
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
| | - Christine A Pratilas
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Emi Caywood
- Division of Hematology/Oncology, Nemours Children's Health, Jacksonville, Florida
| | - Jonathan Metts
- Sarcoma Department, Moffitt Cancer Center, Tampa, Florida
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Peterburg, Florida
| | - Hong Yin
- Department of Pathology, Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Brooke Fridley
- Health Services and Outcomes Research, Children's Mercy Hospital, Kansas City, Missouri
| | - Jun Yin
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Jose Laborde
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Damon R Reed
- Moffitt Cancer Center, Department of Individualized Cancer Management, Tampa, Florida
| | | | - Lars M Wagner
- Division of Pediatric Hematology/Oncology, Duke University, Durham, North Carolina
| |
Collapse
|
2
|
Xiao J, Zhao F, Luo W, Yang G, Wang Y, Qiu J, Liu Y, You L, Zheng L, Zhang T. Human Equilibrative Nucleoside Transporter 1: Novel Biomarker and Prognostic Indicator for Patients with Gemcitabine-Treated Pancreatic Cancer. Cancer Manag Res 2024; 16:651-661. [PMID: 38919872 PMCID: PMC11198018 DOI: 10.2147/cmar.s465098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Aim This article aimed to find appropriate pancreatic cancer (PC) patients to treat with Gemcitabine with better survival outcomes by detecting hENT1 levels. Methods We collected surgical pathological tissues from PC patients who received radical surgery in our hospital from September 2004 to December 2014. A total of 375 PC tissues and paired adjacent nontumor tissues were employed for the construction of 4 tissue microarrays (TMAs). The quality of the 4 TMAs was examined by HE staining. We performed immunohistochemistry analysis to evaluate hENT1 expression in the TMAs. Moreover, we detected hENT1 expression level and proved the role of hENT1 in cell proliferation, drug resistance, migration and invasion in vivo and vitro. Results The results indicated that low hENT1 expression indicated a significantly poor outcome in PC patients, including shortened DFS (21.6±2.8 months versus 36.9±4.0 months, p<0.001) and OS (33.6±3.9 versus 39.6±3.9, p=0.004). Meanwhile, patients in stage I/II of TNM stage had a longer OS (40.2±3.4 versus 15.4±1.7, p=0.002) and DFS (31.0±3.1 versus 12.4±1.9, p=0.016) than patients in stage III/IV. Patients in M0 stage had a longer OS (39.7±3.4 versus 16.2±1.9, p=0.026) and DFS(30.7±3.0 versus 11.8±2.2, p=0.031) than patients in M1 stage, and patients with tumors not invading the capsule had a better DFS than those with tumor invasion into the capsule (30.8±3.0 versus 12.6±2.3, p=0.053). Patients with preoperative CA19-9 values ≤467 U/mL have longer DFS than that of patients who had preoperative CA19-9 values >467 U/mL (37.9±4.1 versus 22.9±4.0, p=0.04). In the subgroup analysis, a high hENT1 expression level was related to a longer OS(39.4±4.0 versus 31.5±3.9, p=0.001) and DFS(35.7±4.0 versus 20.6±2.7; p<0.0001) in the Gemcitabine subgroup. Conclusion PC patients with high hENT1 expression have a better survival outcomes when receiving Gemcitabine. hENT1 expression can be a great prognostic indicator for PC patients to receive Gemcitabine treatment.
Collapse
Affiliation(s)
- Jianchun Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Fangyu Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Wenhao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Yicheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Jiangdong Qiu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Yueze Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
- Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, People’s Republic of China
| |
Collapse
|
3
|
Denu RA, Dann AM, Keung EZ, Nakazawa MS, Nassif Haddad EF. The Future of Targeted Therapy for Leiomyosarcoma. Cancers (Basel) 2024; 16:938. [PMID: 38473300 PMCID: PMC10930698 DOI: 10.3390/cancers16050938] [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: 01/18/2024] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Leiomyosarcoma (LMS) is an aggressive subtype of soft tissue sarcoma that arises from smooth muscle cells, most commonly in the uterus and retroperitoneum. LMS is a heterogeneous disease with diverse clinical and molecular characteristics that have yet to be fully understood. Molecular profiling has uncovered possible targets amenable to treatment, though this has yet to translate into approved targeted therapies in LMS. This review will explore historic and recent findings from molecular profiling, highlight promising avenues of current investigation, and suggest possible future strategies to move toward the goal of molecularly matched treatment of LMS. We focus on targeting the DNA damage response, the macrophage-rich micro-environment, the PI3K/mTOR pathway, epigenetic regulators, and telomere biology.
Collapse
Affiliation(s)
- Ryan A. Denu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Amanda M. Dann
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Emily Z. Keung
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Michael S. Nakazawa
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elise F. Nassif Haddad
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
4
|
Persaud AK, Bernier MC, Massey MA, Agrawal S, Kaur T, Nayak D, Xie Z, Weadick B, Raj R, Hill K, Abbott N, Joshi A, Anabtawi N, Bryant C, Somogyi A, Cruz-Monserrate Z, Amari F, Coppola V, Sparreboom A, Baker SD, Unadkat JD, Phelps MA, Govindarajan R. Increased renal elimination of endogenous and synthetic pyrimidine nucleosides in concentrative nucleoside transporter 1 deficient mice. Nat Commun 2023; 14:3175. [PMID: 37264059 PMCID: PMC10235067 DOI: 10.1038/s41467-023-38789-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Concentrative nucleoside transporters (CNTs) are active nucleoside influx systems, but their in vivo roles are poorly defined. By generating CNT1 knockout (KO) mice, here we identify a role of CNT1 in the renal reabsorption of nucleosides. Deletion of CNT1 in mice increases the urinary excretion of endogenous pyrimidine nucleosides with compensatory alterations in purine nucleoside metabolism. In addition, CNT1 KO mice exhibits high urinary excretion of the nucleoside analog gemcitabine (dFdC), which results in poor tumor growth control in CNT1 KO mice harboring syngeneic pancreatic tumors. Interestingly, increasing the dFdC dose to attain an area under the concentration-time curve level equivalent to that achieved by wild-type (WT) mice rescues antitumor efficacy. The findings provide new insights into how CNT1 regulates reabsorption of endogenous and synthetic nucleosides in murine kidneys and suggest that the functional status of CNTs may account for the optimal action of pyrimidine nucleoside analog therapeutics in humans.
Collapse
Affiliation(s)
- Avinash K Persaud
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Matthew C Bernier
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael A Massey
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Life Sciences Education, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Shipra Agrawal
- Division of Nephrology & Hypertension, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tejinder Kaur
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Debasis Nayak
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhiliang Xie
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Brenna Weadick
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Kasey Hill
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Nicole Abbott
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Arnav Joshi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Nadeen Anabtawi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Claire Bryant
- Center for Clinical & Translational Research, Nationwide Children's Hospital, Columbus, OH, 43210, USA
| | - Arpad Somogyi
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Foued Amari
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Alex Sparreboom
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharyn D Baker
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, College of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Mitch A Phelps
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA.
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
5
|
Nayak D, Weadick B, Govindarajan R. Combination of Tissue Microarray Profiling and Multiplexed IHC Approaches to Investigate Transport Mechanism of Nucleoside Analog Drug Resistance. Methods Mol Biol 2023; 2660:95-121. [PMID: 37191793 PMCID: PMC10311792 DOI: 10.1007/978-1-0716-3163-8_8] [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: 05/17/2023]
Abstract
Nucleoside analogs (NAs) are an established class of anticancer agents being used clinically for the treatment of diverse cancers, either as monotherapy or in combination with other established anticancer or pharmacological agents. To date, nearly a dozen anticancer NAs are approved by the FDA, and several novel NAs are being tested in preclinical and clinical trials for future applications. However, improper delivery of NAs into tumor cells because of alterations in expression of one or more drug carrier proteins (e.g., solute carrier (SLC) transporters) within tumor cells or cells surrounding the tumor microenvironment stands as one of the primary reasons for therapeutic drug resistance. The combination of tissue microarray (TMA) and multiplexed immunohistochemistry (IHC) is an advanced, high-throughput approach over conventional IHC that enables researchers to effectively investigate alterations to numerous such chemosensitivity determinants simultaneously in hundreds of tumor tissues derived from patients. In this chapter, taking an example of a TMA from pancreatic cancer patients treated with gemcitabine (a NA chemotherapeutic agent), we describe the step-by-step procedure of performing multiplexed IHC, imaging of TMA slides, and quantification of expression of some relevant markers in these tissue sections as optimized in our laboratory and discuss considerations while designing and carrying out this experiment.
Collapse
Affiliation(s)
- Debasis Nayak
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - Brenna Weadick
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics and Pharmacology, The Ohio State University College of Pharmacy, Columbus, OH, USA.
- Translational Therapeutics, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
6
|
Lin Q, Shen S, Qian Z, Rasam SS, Serratore A, Jusko WJ, Kandel ES, Qu J, Straubinger RM. Comparative Proteomic Analysis Identifies Key Metabolic Regulators of Gemcitabine Resistance in Pancreatic Cancer. Mol Cell Proteomics 2022; 21:100409. [PMID: 36084875 PMCID: PMC9582795 DOI: 10.1016/j.mcpro.2022.100409] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 08/21/2022] [Accepted: 09/04/2022] [Indexed: 01/18/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is highly refractory to treatment. Standard-of-care gemcitabine (Gem) provides only modest survival benefits, and development of Gem resistance (GemR) compromises its efficacy. Highly GemR clones of Gem-sensitive MIAPaCa-2 cells were developed to investigate the molecular mechanisms of GemR and implemented global quantitative differential proteomics analysis with a comprehensive, reproducible ion-current-based MS1 workflow to quantify ∼6000 proteins in all samples. In GemR clone MIA-GR8, cellular metabolism, proliferation, migration, and 'drug response' mechanisms were the predominant biological processes altered, consistent with cell phenotypic alterations in cell cycle and motility. S100 calcium binding protein A4 was the most downregulated protein, as were proteins associated with glycolytic and oxidative energy production. Both responses would reduce tumor proliferation. Upregulation of mesenchymal markers was prominent, and cellular invasiveness increased. Key enzymes in Gem metabolism pathways were altered such that intracellular utilization of Gem would decrease. Ribonucleoside-diphosphate reductase large subunit was the most elevated Gem metabolizing protein, supporting its critical role in GemR. Lower Ribonucleoside-diphosphate reductase large subunit expression is associated with better clinical outcomes in PDAC, and its downregulation paralleled reduced MIAPaCa-2 proliferation and migration and increased Gem sensitivity. Temporal protein-level Gem responses of MIAPaCa-2 versus GemR cell lines (intrinsically GemR PANC-1 and acquired GemR MIA-GR8) implicate adaptive changes in cellular response systems for cell proliferation and drug transport and metabolism, which reduce cytotoxic Gem metabolites, in DNA repair, and additional responses, as key contributors to the complexity of GemR in PDAC. These findings additionally suggest targetable therapeutic vulnerabilities for GemR PDAC patients.
Collapse
Affiliation(s)
- Qingxiang Lin
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Zhicheng Qian
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Sailee S Rasam
- Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Andrea Serratore
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA
| | - Eugene S Kandel
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jun Qu
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, New York, USA.
| | - Robert M Straubinger
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA; Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, USA; Center of Excellence in Bioinformatics & Life Science, University at Buffalo, State University of New York, Buffalo, New York, USA; Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA.
| |
Collapse
|
7
|
Watson S, Verret B, Ropert S, Adam J, Bahleda R, Briand S, Cavalcanti A, Chamseddine AN, Court C, Fadel E, Faron M, Haddag‐Miliani L, Henon C, Pechoux CL, Levy A, Mercier O, Ngo C, Honoré C, Cesne AL, Mir O. Single-agent gemcitabine in patients with advanced, pre-treated angiosarcoma: A multicenter, retrospective study. Cancer Med 2022; 12:3160-3166. [PMID: 35971325 PMCID: PMC9939156 DOI: 10.1002/cam4.5147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/16/2022] [Accepted: 08/02/2022] [Indexed: 11/08/2022] Open
Abstract
Gemcitabine has shown clinical activity against angiosarcoma in small series, alone, or combined with taxanes. We aimed to evaluate its activity as a single-agent in a larger series of patients with advanced angiosarcoma. We retrospectively reviewed the electronic medical records of consecutive adult patients with advanced angiosarcoma treated with single-agent gemcitabine at our institutions from January 2010 to January 2021. Response was evaluated according to RECIST 1.1, and toxicity was graded according to NCI-CTC v5.0. 42 patients were identified. 38 patients (90%) had received prior anthracyclines and weekly paclitaxel, and 9 (21%) had received pazopanib. The best tumor response was partial response (PR) in 16 patients (38%), or stable disease (10 patients, 24%). All 8 patients with cardiac angiosarcoma experienced a PR. Median PFS was 5.4 months (95%CI: 3.1-6.5), and median OS was 9.9 months (95%CI: 6.6-13.4). Single-agent gemcitabine has clinically meaningful activity in advanced, heavily pre-treated angiosarcoma.
Collapse
Affiliation(s)
- Sarah Watson
- Department of Medical OncologyCurie InstituteParisFrance
| | | | - Stanislas Ropert
- Department of Medical OncologyAntony Private HospitalAntonyFrance
| | - Julien Adam
- Division of Biology and PathologyGustave RoussyVillejuifFrance
| | | | - Sylvain Briand
- Department of Orthopedic Surgery, Kremlin‐Bicêtre Teaching HospitalUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | | | | | - Charles Court
- Department of Orthopedic Surgery, Kremlin‐Bicêtre Teaching HospitalUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Elie Fadel
- Department of Thoracic Surgery, Marie Lannelongue Teaching HospitalUniversité Paris‐SaclayLe Plessis‐RobinsonFrance
| | | | | | - Clémence Henon
- Division of Cancer MedicineGustave RoussyVillejuifFrance
| | | | - Antonin Levy
- Division of Radiation OncologyGustave RoussyVillejuifFrance
| | - Olaf Mercier
- Department of Thoracic Surgery, Marie Lannelongue Teaching HospitalUniversité Paris‐SaclayLe Plessis‐RobinsonFrance
| | - Carine Ngo
- Division of Biology and PathologyGustave RoussyVillejuifFrance
| | | | - Axel Le Cesne
- Division of International Patients CareGustave RoussyVillejuifFrance
| | - Olivier Mir
- Division of Cancer MedicineGustave RoussyVillejuifFrance,Department of Ambulatory Cancer CareGustave RoussyVillejuifFrance
| |
Collapse
|
8
|
Elisabetta G, Anna B, Adriano P, Andrea CD, Guido S, Ilaria P, Andrea B, Lorenzo A, Serena P. Pharmacogenomics of soft tissue sarcomas: New horizons to understand efficacy and toxicity. Cancer Treat Res Commun 2022; 31:100528. [PMID: 35123198 DOI: 10.1016/j.ctarc.2022.100528] [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: 10/13/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023]
Abstract
Clinical responses to anticancer therapies in advanced soft tissue sarcoma (STS) are unfortunately limited to a small subset of patients. Much of the inter-individual variability in treatment efficacy and risk of toxicities is as result of polymorphisms in genes encoding proteins involved in drug pharmacokinetics and pharmacodynamics. Therefore, the detection of pharmacogenomics (PGx) biomarkers that might predict drug response and toxicity can be useful to explain the genetic basis for the differences in treatment efficacy and toxicity among STS patients. PGx markers are frequently located in transporters, drug-metabolizing enzyme genes, drug targets, or HLA alleles. Along this line, genetic variability harbouring in the germline genome of the patients can influence systemic pharmacokinetics and pharmacodynamics of the treatments, acting as predictive biomarkers for drug-induced toxicity and treatment efficacy. By linking drug activity to the functional complexity of cancer genomes, also systematic pharmacogenomic profiling in cancer cell lines and primary STS samples represents area of active investigation that could eventually lead to enhanced efficacy and offer a powerful biomarker discovery platform to optimize current treatments and improve the knowledge about the individual's drug response in STS patients into the clinical practice.
Collapse
Affiliation(s)
| | - Boddi Anna
- Department of Health Science, University of Florence, Florence, Italy
| | - Pasqui Adriano
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Campanacci Domenico Andrea
- Department of Health Science, University of Florence, Florence, Italy; Department of Orthopaedic Oncology and Reconstructive Surgery, Careggi University Hospital, Florence, Italy
| | - Scoccianti Guido
- Department of Health Science, University of Florence, Florence, Italy
| | - Palchetti Ilaria
- Department of Chemistry Ugo Schiff, University of Florence, Sesto Fiorentino, Florence, Italy
| | - Bernini Andrea
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, Siena, 53100 Italy
| | - Antonuzzo Lorenzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy; Medical Oncology Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pillozzi Serena
- Medical Oncology Unit, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
9
|
Zhao X, Ma Y, Dong X, Zhang Z, Tian X, Zhao X, Yang Y. Molecular characterization of circulating tumor cells in pancreatic ductal adenocarcinoma: potential diagnostic and prognostic significance in clinical practice. Hepatobiliary Surg Nutr 2021; 10:796-810. [PMID: 35004946 DOI: 10.21037/hbsn-20-383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/07/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND The clinical value of heterogeneous sub-populations of circulating tumor cells (CTCs) in pancreatic ductal adenocarcinoma (PDAC) remains unclear. METHODS Peripheral blood samples were obtained from 67 PDAC patients. CTCs were isolated by employing CD45 negative enrichment technique and further characterized for epithelial to mesenchymal transition (EMT) or human equilibrative nucleoside transporter-1 (hENT-1). The relationships between CTCs sub-phenotypes with clinicopathological factors or post-operative recurrence in PDAC patients were analyzed. RESULTS EMT related CTCs could be isolated and identified from the 81% of patients (54/67), and both the total count (median: 5 vs. 17/mL, P<0.0001) and M-CTC percentage (median: 0.2 vs. 0.345, P=0.0244) of CTCs could differentiate local/regional with metastatic disease. Multivariate analysis showed that both AJCC stage (P=0.025) and M-CTC percentage (P=0.001) were independent prognostic indicators of recurrence free survival (RFS) in resected patients. Moreover, Kaplan-Meier survival analysis showed that M-CTC after 2 courses of chemotherapy was significantly associated with inferior RFS (49.5 weeks vs. undefined, P=0.0288). No significant correlation in hENT-1 expression was found between CTCs and matched tumor tissues, and further multivariate analysis suggested hENT-1 expression in CTCs as independent prognostic factor for RFS (P=0.016). Patients with low hENT-1 expression in CTCs had decreased RFS (32 weeks vs. undefined, P=0.0337). CONCLUSIONS CTCs could be the promising diagnostic biomarkers in PDAC patients, and phenotypic profiling of CTCs based on EMT or hENT-1 could help establish novel prognostic biomarkers in resected patients undergoing adjuvant gemcitabine-based chemotherapy. KEYWORDS Circulating tumor cells (CTCs); Pancreatic ductal adenocarcinoma (PDAC); Epithelial to mesenchymal transition (EMT); human equilibrative nucleoside transporter-1 (hENT-1).
Collapse
Affiliation(s)
- Xudong Zhao
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yongsu Ma
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Xiu Dong
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhengkui Zhang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
10
|
Carter CJ, Mekkawy AH, Morris DL. Role of human nucleoside transporters in pancreatic cancer and chemoresistance. World J Gastroenterol 2021; 27:6844-6860. [PMID: 34790010 PMCID: PMC8567477 DOI: 10.3748/wjg.v27.i40.6844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/19/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023] Open
Abstract
The prognosis of pancreatic cancer is poor with the overall 5-year survival rate of less than 5% changing minimally over the past decades and future projections predicting it developing into the second leading cause of cancer related mortality within the next decade. Investigations into the mechanisms of pancreatic cancer development, progression and acquired chemoresistance have been constant for the past few decades, thus resulting in the identification of human nucleoside transporters and factors affecting cytotoxic uptake via said transporters. This review summaries the aberrant expression and role of human nucleoside transports in pancreatic cancer, more specifically human equilibrative nucleoside transporter 1/2 (hENT1, hENT2), and human concentrative nucleoside transporter 1/3 (hCNT1, hCNT3), while briefly discussing the connection and importance between these nucleoside transporters and mucins that have also been identified as being aberrantly expressed in pancreatic cancer. The review also discusses the incidence, current diagnostic techniques as well as the current therapeutic treatments for pancreatic cancer. Furthermore, we address the importance of chemoresistance in nucleoside analogue drugs, in particular, gemcitabine and we discuss prospective therapeutic treatments and strategies for overcoming acquired chemoresistance in pancreatic cancer by the enhancement of human nucleoside transporters as well as the potential targeting of mucins using a combination of mucolytic compounds with cytotoxic agents.
Collapse
Affiliation(s)
- Carly Jade Carter
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, St George Hospital, University of New South Wales, Sydney 2217, New South Wales, Australia
- Mucpharm Pty Ltd, Australia
| | - Ahmed H Mekkawy
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, St George Hospital, University of New South Wales, Sydney 2217, New South Wales, Australia
- Mucpharm Pty Ltd, Australia
| | - David L Morris
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, St George Hospital, University of New South Wales, Sydney 2217, New South Wales, Australia
- Mucpharm Pty Ltd, Australia
| |
Collapse
|
11
|
The Prognostic Significance of the BIN1 and CCND2 Gene in Adult Patients with Acute Myeloid Leukemia. Indian J Hematol Blood Transfus 2021; 38:481-491. [DOI: 10.1007/s12288-021-01479-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/28/2021] [Indexed: 10/20/2022] Open
|
12
|
Martínez-Trufero J, Cruz Jurado J, Gómez-Mateo MC, Bernabeu D, Floría LJ, Lavernia J, Sebio A, García Del Muro X, Álvarez R, Correa R, Hernández-León CN, Marquina G, Hindi N, Redondo A, Martínez V, Asencio JM, Mata C, Valverde Morales CM, Martin-Broto J. Uncommon and peculiar soft tissue sarcomas: Multidisciplinary review and practical recommendations for diagnosis and treatment. Spanish group for Sarcoma research (GEIS - GROUP). Part I. Cancer Treat Rev 2021; 99:102259. [PMID: 34311246 DOI: 10.1016/j.ctrv.2021.102259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 12/22/2022]
Affiliation(s)
| | - Josefina Cruz Jurado
- Hospital Universitario Canarias, Medical Oncology Department, Santa Cruz de Tenerife, Spain
| | | | - Daniel Bernabeu
- Hospital Universitario La Paz, Radiology Department, Madrid, Spain
| | - Luis Javier Floría
- Hospital Universitario Miguel Servet, Orthopedic and Traumatology Department, Zaragoza, Spain
| | - Javier Lavernia
- Instituto Valenciano de Oncología, Medical Oncology Department, Valencia, Spain
| | - Ana Sebio
- Hospital Universitario Santa Creu i Sant Pau, Medical Oncology Department, Barcelona, Spain
| | | | - Rosa Álvarez
- Hospital Universitario Gregorio Marañón, Medical Oncology Department, Madrid, Spain
| | - Raquel Correa
- Hospital Virgen de la Victoria, Radiation Oncology Department, Malaga, Spain
| | | | - Gloria Marquina
- Hospital Universitario Clínico San Carlos, Medical Oncology Department, Madrid, Spain
| | - Nadia Hindi
- University Hospital "Fundacion Jimenez Diaz" Madrid, Medical Oncology Department, Madrid, Research Institute FJD-UAM, Madrid (Spain), TBsarc, CITIUS III, Seville, Spain
| | - Andrés Redondo
- Hospital Universitario La Paz, Medical Oncology Department, Madrid, Spain
| | - Virginia Martínez
- Hospital Universitario La Paz, Medical Oncology Department, Madrid, Spain
| | | | - Cristina Mata
- Hospital Universitario Gregorio Marañón, Pediatric and Adolescent Hemato-oncology Department, Madrid, Spain
| | | | - Javier Martin-Broto
- University Hospital "Fundacion Jimenez Diaz" Madrid, Medical Oncology Department, Madrid, Research Institute FJD-UAM, Madrid (Spain), TBsarc, CITIUS III, Seville, Spain
| |
Collapse
|
13
|
Akrivi S, Varras M, Anastasiadi Z, Pappa C, Vlachioti A, Varra VK, Varra FN, Balasi E, Akrivis C. Primary vulvar leiomyosarcoma localized in the Bartholin's gland area: A case report and review. Mol Clin Oncol 2021; 14:69. [PMID: 33680460 PMCID: PMC7890440 DOI: 10.3892/mco.2021.2231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 01/14/2021] [Indexed: 01/14/2023] Open
Abstract
Vulvar sarcomas located in the Bartholin's gland area are extremely uncommon mesenchymal vulvar tumors. These neoplasms can be mistaken as Bartholin' gland benign lesions such as cysts or abscesses, leading to a delay in the diagnosis of underlying malignancy. Currently, only a few cases of these aggressive cancers have been reported in the literature. A 42-year-old female patient without any previous complaint presented to Obstetrics and Gynecology Department of ‘G. Chaztikosta’ General Hospital due to a vulvar lump in the area of the left Bartholin's gland with a 6-month history of progressive swelling. Pelvic examination showed a solid mass of 6.5-cm in maximum diameter, localized in the left Bartholin's gland. The patient underwent wide local excision and histopathological examination of hematoxylin and eosin-stained sections indicated intersecting fascicles of spindle cells, with moderate to severe atypia. The number of mitoses was up to 8 per 10 high power fields. The neoplasm to its greatest extent was circumscribed and in places had an invasive growth pattern. Tumoral necrosis was not seen. Involved Bartholin' gland by the tumor was identified. The tumor extended focally to the surgical margin. The neoplastic cells showed positive staining for smooth muscle actin, desmin, HHF35, caldesmon, vimentin and estrogen and progesterone receptors. Immunohistochemistry was negative for S100, myoglobulin, keratin 116, CD117, CD34 and CD31. The patient denied further surgery or/and local radiotherapy, although the mass was >5-cm and a focally infiltrative surgical margin was found. During the close follow-up, no local recurrences or metastases were observed 53 months after surgery. In conclusion, wide local tumor excision with free surgical margins is a good option of surgery for vulvar leiomyosarcomas. In recurrences, a new extensive surgical resection of the lesion and radiotherapy are suggested. Ipsilateral lympadenectomy is indicated when there is a pathologic lymph node. Chemotherapy is provided in cases of distal metastases.
Collapse
Affiliation(s)
- Stella Akrivi
- Department of Obstetrics and Gynecology, Royal Jubille Maternity Hospital, Belfast Trust, Belfast BT12 6BA, UK
| | - Michail Varras
- Fourth Department of Obstetrics and Gynecology, 'Elena Venizelou' General Hospital, Athens 11521, Greece
| | - Zoi Anastasiadi
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | - Christina Pappa
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | - Aikaterini Vlachioti
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | | | - Fani-Niki Varra
- Department of Pharmacy, Frederick University, Nicosia 1036, Cyprus
| | - Eufemia Balasi
- Pathology Department, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| | - Christos Akrivis
- Department of Obstetrics and Gynecology, 'G. Chatzikosta' General Hospital, Ioannina 45001, Epirus, Greece
| |
Collapse
|
14
|
Miller SR, Zhang X, Hau RK, Jilek JL, Jennings EQ, Galligan JJ, Foil DH, Zorn KM, Ekins S, Wright SH, Cherrington NJ. Predicting Drug Interactions with Human Equilibrative Nucleoside Transporters 1 and 2 Using Functional Knockout Cell Lines and Bayesian Modeling. Mol Pharmacol 2021; 99:147-162. [PMID: 33262250 PMCID: PMC7816041 DOI: 10.1124/molpharm.120.000169] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Equilibrative nucleoside transporters (ENTs) 1 and 2 facilitate nucleoside transport across the blood-testis barrier (BTB). Improving drug entry into the testes with drugs that use endogenous transport pathways may lead to more effective treatments for diseases within the reproductive tract. In this study, CRISPR/CRISPR-associated protein 9 was used to generate HeLa cell lines in which ENT expression was limited to ENT1 or ENT2. We characterized uridine transport in these cell lines and generated Bayesian models to predict interactions with the ENTs. Quantification of [3H]uridine uptake in the presence of the ENT-specific inhibitor S-(4-nitrobenzyl)-6-thioinosine (NBMPR) demonstrated functional loss of each transporter. Nine nucleoside reverse-transcriptase inhibitors and 37 nucleoside/heterocycle analogs were evaluated to identify ENT interactions. Twenty-one compounds inhibited uridine uptake and abacavir, nevirapine, ticagrelor, and uridine triacetate had different IC50 values for ENT1 and ENT2. Total accumulation of four identified inhibitors was measured with and without NBMPR to determine whether there was ENT-mediated transport. Clofarabine and cladribine were ENT1 and ENT2 substrates, whereas nevirapine and lexibulin were ENT1 and ENT2 nontransported inhibitors. Bayesian models generated using Assay Central machine learning software yielded reasonably high internal validation performance (receiver operator characteristic > 0.7). ENT1 IC50-based models were generated from ChEMBL; subvalidations using this training data set correctly predicted 58% of inhibitors when analyzing activity by percent uptake and 63% when using estimated-IC50 values. Determining drug interactions with these transporters can be useful in identifying and predicting compounds that are ENT1 and ENT2 substrates and can thereby circumvent the BTB through this transepithelial transport pathway in Sertoli cells. SIGNIFICANCE STATEMENT: This study is the first to predict drug interactions with equilibrative nucleoside transporter (ENT) 1 and ENT2 using Bayesian modeling. Novel CRISPR/CRISPR-associated protein 9 functional knockouts of ENT1 and ENT2 in HeLa S3 cells were generated and characterized. Determining drug interactions with these transporters can be useful in identifying and predicting compounds that are ENT1 and ENT2 substrates and can circumvent the blood-testis barrier through this transepithelial transport pathway in Sertoli cells.
Collapse
Affiliation(s)
- Siennah R Miller
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Xiaohong Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Raymond K Hau
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Joseph L Jilek
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Erin Q Jennings
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - James J Galligan
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Daniel H Foil
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Kimberley M Zorn
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Sean Ekins
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Stephen H Wright
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| | - Nathan J Cherrington
- Department of Pharmacology and Toxicology, College of Pharmacy (S.R.M., R.K.H., J.L.J., E.Q.J., J.J.G., N.J.C.), and Department of Physiology, College of Medicine (X.Z., S.H.W.), University of Arizona, Tucson, Arizona and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (D.H.F., K.M.Z., S.E.)
| |
Collapse
|
15
|
Noguchi S, Takagi A, Tanaka T, Takahashi Y, Pan X, Kibayashi Y, Mizokami R, Nishimura T, Tomi M. Fluorouracil uptake in triple-negative breast cancer cells: Negligible contribution of equilibrative nucleoside transporters 1 and 2. Biopharm Drug Dispos 2021; 42:85-93. [PMID: 33426680 DOI: 10.1002/bdd.2261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 12/15/2020] [Accepted: 12/20/2020] [Indexed: 01/07/2023]
Abstract
Equilibrative nucleoside transporters (ENTs) 1 and 2 reportedly accept fluorouracil as a substrate. Here, we evaluated ENT1/2 expression at the messenger RNA (mRNA), protein, and functional levels in a panel of four triple-negative breast cancer (TNBC) cell lines, BT-549, Hs578T, MDA-MB-231, and MDA-MB-435, and we examined the relationship of the observed profiles to fluorouracil sensitivity. Nitrobenzylthioinosine (NBMPR) at 0.1 μM inhibits only ENT1, while dipyridamole at 10 μM or NBMPR at 100 μM inhibits both ENT1 and ENT2. We found that the uptake of [3 H]uridine, a typical substrate of ENT1 and ENT2, was decreased to approximately 40% by 0.1 μM NBMPR. At 100 μM, NBMPR almost completely blocked the saturable uptake of [3 H]uridine, but this does not imply a functional role of ENT2, because 10 μM dipyridamole showed similar inhibition to 0.1 μM NBMPR. Expression of ENT1 mRNA was almost 1 order of magnitude higher than that of ENT2 in all TNBC cell lines. Liquid chromatography-tandem mass spectrometry(LC-MS/MS) LC-MS/MS-based targeted protein quantification showed that ENT1 protein levels were in the range of 9.3-30 fmol/μg protein in plasma membrane fraction of TNBC cell lines, whereas ENT2 protein was below the detection limit. [3 H]Fluorouracil uptake was insensitive to 0.1 μM NBMPR and 10 μM dipyridamole, suggesting a negligible contribution of ENT1 and ENT2 to fluorouracil uptake. The levels of ENT1 mRNA, ENT1 protein, ENT2 mRNA, and ENT1-mediated [3 H]uridine uptake in the four TNBC cell lines showed no correlation with fluorouracil sensitivity. These results indicate that neither ENT1 nor ENT2 contributes significantly to the fluorouracil sensitivity of TNBC cell lines.
Collapse
Affiliation(s)
- Saki Noguchi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Akinori Takagi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan.,Laboratory of Applied Therapeutics, Showa Pharmaceutical University, Tokyo, Japan
| | - Takahiro Tanaka
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Yu Takahashi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Xiaole Pan
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Yuka Kibayashi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Ryo Mizokami
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Tomohiro Nishimura
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Masatoshi Tomi
- Division of Pharmaceutics, Faculty of Pharmacy, Keio University, Tokyo, Japan
| |
Collapse
|
16
|
Genomic Database Analysis of Uterine Leiomyosarcoma Mutational Profile. Cancers (Basel) 2020; 12:cancers12082126. [PMID: 32751892 PMCID: PMC7464219 DOI: 10.3390/cancers12082126] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/14/2022] Open
Abstract
Uterine Leiomyosarcoma (uLMS) is by far the most common type of uterine sarcoma, characterized by an aggressive clinical course, a heterogeneous genetic profile and a very scarce response to cytotoxic chemotherapy. The genetic make-up of uLMS is an area of active study that could provide essential cues for the development of new therapeutic approaches. A total of 216 patients with uLMS from cBioPortal and AACR-GENIE databases were included in the study. The vast majority of patients (81%) carried at least one mutation in either TP53, RB1, ATRX or PTEN. The most frequently mutated gene was TP53, with 61% of the patients harboring at least one mutation, followed by RB1 at 48%. PTEN alteration was more frequent in metastases than in primary lesions, consistent with a later acquisition during tumor progression. There was a significant trend for TP53 and RB1 mutations to occur together, while both TP53 and RB1 were mutually exclusive with respect to CDKN2A/B inactivation. Overall survival did not show significant correlation with the mutational status, even if RB1 mutation emerged as a favorable prognostic factor in the TP53-mutant subgroup. This comprehensive analysis shows that uLMS is driven almost exclusively by the inactivation of tumor suppressor genes and suggests that future therapeutic strategies should be directed at targeting the main genetic drivers of uLMS oncogenesis.
Collapse
|
17
|
Impact of Chemotherapy in the Adjuvant Setting of Early Stage Uterine Leiomyosarcoma: A Systematic Review and Updated Meta-Analysis. Cancers (Basel) 2020; 12:cancers12071899. [PMID: 32674439 PMCID: PMC7409135 DOI: 10.3390/cancers12071899] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/08/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Although the use of adjuvant chemotherapy (AC) appears to be increasing over the past few years, several clinical trials and previous meta-analyses failed to determine whether AC could improve clinical outcomes in uterine leiomyosarcoma (uLMS). The aim of this systematic review and meta-analysis was to compare AC (with or without radiotherapy) versus observation (obs) after primary surgery in early stage uLMS. Materials and Methods: Randomized controlled (RCTs) and non-randomized studies (NRSs) were retrieved. Outcomes of interest were as follows: distant recurrence rate, locoregional recurrence rate and overall recurrence rate. Results about distant recurrence rate, locoregional recurrence rate and overall recurrence rate were compared by calculating odds ratios (ORs) with 95% confidence intervals (CIs); ORs were combined with Mantel–Haenszel method. Results: Nine studies were included in the analysis, involving 545 patients (AC: 252, obs: 293). Compared with obs, AC did not reduce locoregional and distant recurrence rate, with a pooled OR of 1.36 and 0.63, respectively. Similarly, administration of AC did not decrease overall recurrence rate in comparison to obs. Conclusion: According to our results, AC (with or without radiotherapy) did not decrease recurrence rate in early stage uLMS; thus, the role of AC in this setting remains unclear.
Collapse
|
18
|
Oesterheld JE, Reed DR, Setty BA, Isakoff MS, Thompson P, Yin H, Hayashi M, Loeb DM, Smith T, Makanji R, Fridley BL, Wagner LM. Phase II trial of gemcitabine and nab-paclitaxel in patients with recurrent Ewing sarcoma: A report from the National Pediatric Cancer Foundation. Pediatr Blood Cancer 2020; 67:e28370. [PMID: 32386107 PMCID: PMC7771264 DOI: 10.1002/pbc.28370] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 04/02/2020] [Accepted: 04/13/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND The combination of gemcitabine and docetaxel is often used to treat patients with recurrent sarcoma. Nab-paclitaxel is a taxane modified to improve drug exposure and increase intratumoral accumulation and, in combination with gemcitabine, is standard therapy for pancreatic cancer. Applying the dosages and schedule used for pancreatic cancer, we performed a phase II trial to assess the response rate of gemcitabine and nab-paclitaxel in patients with relapsed Ewing sarcoma. PROCEDURE Using a Simon's two-stage design to identify a response rate of ≥ 35%, patients received nab-paclitaxel 125 mg/m2 followed by gemcitabine 1000 mg/m2 i.v. on days 1, 8, and 15 of four-week cycles. Immunohistochemical analysis of archival tissue was performed to identify possible biomarkers of response. RESULTS Eleven patients from four institutions enrolled, with a median age of 22 years (range, 14-27). Patients were heavily pretreated (median 3 prior regimens, range, 1-7). Thirty-five cycles were administered (median 2, range, 1-8). Accrual was stopped after 11 patients, due to only one confirmed partial response. Two other patients had partial responses after two cycles, but withdrew because of adverse effects or progression before confirmation of continued response. The predominant toxicity was myelosuppression, and four (36%) patients were removed due to hematologic toxicity despite pegfilgrastim and dose reductions. Expression of secreted protein, acidic and rich in cysteine (SPARC) and CAV-1 in archival tumors was not predictive of clinical benefit in this small cohort of patients. CONCLUSIONS In patients with heavily pretreated Ewing sarcoma, the confirmed response rate of 9% was similar to multi-institutional studies of gemcitabine and docetaxel.
Collapse
Affiliation(s)
- Javier E. Oesterheld
- Department of Pediatric Hematology, Oncology, Bone Marrow Transplantation, and Palliative Care, Levine Children’s Hospital at Atrium Health, Charlotte, North Carolina
| | - Damon R. Reed
- Department of Interdisciplinary Cancer Management, Moffitt Cancer Center Adolescent and Young Adult Program, Tampa, Florida
| | - Bhuvana A. Setty
- Division of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Nationwide Children’s Hospital, Columbus, Ohio
| | - Michael S. Isakoff
- Center for Cancer and Blood Disorders, Connecticut Children’s Medical Center, Hartford, Connecticut
| | - Patrick Thompson
- Division of Pediatric Hematology-Oncology, University of North Carolina Health Care, Chapel Hill, North Carolina
| | - Hong Yin
- Department of Pathology, Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - Masanori Hayashi
- Department of Pediatrics Hematology-Oncology and Bone Marrow Transplant, Children’s Hospital Colorado, Aurora, Colorado
| | - David M. Loeb
- Department of Pediatrics and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York City, New York
| | - Tiffany Smith
- National Pediatric Cancer Foundation, Tampa, Florida
| | - Rikesh Makanji
- Department of Diagnostic Imaging and Interventional Radiology, Moffitt Cancer Center, Tampa, Florida
| | - Brooke L. Fridley
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida
| | - Lars M. Wagner
- Division of Pediatric Hematology-Oncology, Duke Children’s Hospital and Health Center, Durham, North Carolina
| |
Collapse
|
19
|
Caruso C, Garofalo C. Pharmacogenomics Biomarkers of Soft Tissue Sarcoma Therapies. Front Oncol 2020; 10:509. [PMID: 32351891 PMCID: PMC7174622 DOI: 10.3389/fonc.2020.00509] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/20/2020] [Indexed: 12/19/2022] Open
Abstract
Soft tissue sarcomas (STS) are heterogeneous rare malignancies comprising ~1% of all solid cancers in adults and including more than 70 histological and molecular subtypes with different pathological and clinical development characteristics. Over the last two decades, the increased knowledge of the new molecular and genomic mechanisms of different STS histotypes allowed for a reclassification of these tumors and consequently to the development of novel chemotherapeutic agents. Generally, surgery, in combination with radiotherapy only in selected cases of localized disease, represents the most common treatment of primary STS, whereas the principal treatment modality for locally advanced or metastatic disease is first-line chemotherapy. The principal treatment for the preponderance of STS patients is usually an anthracycline (epirubicin and doxorubicin) in monotherapy or in combination with other drug novel chemotherapeutic agents. However, survival for treated patients with metastatic disease is poor, and a 2-years survival rate is about 30%. In this scenario, Pharmacogenomics (PGx) biomarkers that can predict drug response play an important role in the improvement of molecular diagnostics in clinical routines and contribute to elucidating the genetic basis for the differences in treatment efficacy and toxicity among STS patients. This review focuses on recent insight in the PGx biomarkers that have been described to modulate responsiveness and toxicity parameters of conventional and new chemotherapeutics drugs in several STS histotypes.
Collapse
Affiliation(s)
| | - Cecilia Garofalo
- Advanced Translational Research Laboratory, Veneto Institute of Oncology IOV – IRCCS, Padua, Italy
| |
Collapse
|
20
|
Akahoshi K, Ban D, Kuboki R, Oba A, Ono H, Mitsunori Y, Kudo A, Tanaka S, Tanabe M. Orotate phosphoribosyltransferase as a predictor of benefit from S-1 adjuvant chemotherapy for cholangiocarcinoma patients. J Gastroenterol Hepatol 2019; 34:1108-1115. [PMID: 30242888 DOI: 10.1111/jgh.14477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/26/2018] [Accepted: 09/12/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIM To improve the prognosis of cholangiocarcinoma, we investigated potential biomarkers that may enable the selection of patients for whom postoperative adjuvant chemotherapy is likely effective. METHODS The cohort of this retrospective study included 170 surgically resected cholangiocarcinoma patients, 26 with gemcitabine adjuvant chemotherapy (GEM group), 36 with S-1 adjuvant chemotherapy (S-1 group), and 103 receiving no adjuvant chemotherapy (NC group). Propensity score matching was performed to adjust patient backgrounds; 36 patients from the NC group then were selected. Immunohistochemistry of orotate phosphoribosyltransferase (OPRT) and human equilibrative nucleoside transporter 1 (hENT1) was performed to determine the correlation between their expression and disease-free survival (DFS). RESULTS After matching, the backgrounds of these three groups were unbiased. No significant improvement of DFS by adjuvant chemotherapy was observed in the whole cohort. However, among the high-OPRT-expression patients, DFS of GEM, S-1, and NC groups at 5 years was 28.8%, 53.8%, and 25.5%, respectively. The DFS of the S-1 group was significantly longer than that of the NC group (P = 0.034). On the other hand, no significant differences in DFS were observed among the low OPRT expression patients. hENT1 expression was shown to have no predictive value. Multivariate analysis of the high-OPRT-expression patients demonstrated that S-1 adjuvant chemotherapy can reduce tumor recurrence (HR, 0.303; P = 0.013). CONCLUSION Cholangiocarcinoma patients with high OPRT expression would benefit from postoperative adjuvant S-1 therapy, which increases the DFS. Assessment of OPRT expression may contribute to the optimization of adjuvant chemotherapy for cholangiocarcinoma.
Collapse
Affiliation(s)
- Keiichi Akahoshi
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryo Kuboki
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Oba
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroaki Ono
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Mitsunori
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Atsushi Kudo
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
21
|
A patient-derived orthotopic xenograft (PDOX) nude-mouse model precisely identifies effective and ineffective therapies for recurrent leiomyosarcoma. Pharmacol Res 2019; 142:169-175. [PMID: 30807865 DOI: 10.1016/j.phrs.2019.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 02/09/2019] [Accepted: 02/22/2019] [Indexed: 01/30/2023]
Abstract
Leiomyosarcoma is a rare and recalcitrant disease. Doxorubicin (DOX) is usually considered first-line treatment for this disease, but frequently is ineffective. In order to individualize therapy for this and other cancers, we have developed the patient-derived orthotopic xenograft (PDOX) mouse model. In the present study, we implanted a recurrent leiomyosarcoma from a resected tumor from the patient's thigh into the femoral muscle of nude mice. The following drugs were tested on the leiomyosarcoma PDOX model: DOX, the combination of gemcitabine (GEM) and docetaxel (DOC), trabectedin (TRA), temozolomide (TEM), pazopanib (PAZ) and olaratumab (OLA). Of these agents GEM/DOC, TRA and TEM were highly effective in the leiomyosarcoma PDOX model, the other agents, including first-line therapy DOX, were ineffective. Thus the leiomyosarcoma PDOX model could precisely distinguish effective and ineffective drugs, demonstrating the potential of the PDOX model for leiomyosarcoma treatment.
Collapse
|