1
|
Salceda-Rivera V, Ortiz-Lazareno PC, Hernández-Flores G, Vazquez-Urrutia JR, Meza-Arroyo J, Pardo-Zepeda M, Romo-Rubio H, Barba-Barba C, Sánchez-Zubieta F, Barrón-Gallardo CA, Gonzalez-Ramella O, Bravo-Cuellar A. Very early remission and increased apoptosis with the use of Pentoxifylline in children with acute lymphoblastic leukemia. Front Oncol 2024; 14:1401262. [PMID: 39421449 PMCID: PMC11484046 DOI: 10.3389/fonc.2024.1401262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Despite the improvement in survival in acute lymphoblastic leukemia (ALL), there are still cases with evasion of chemotherapy-induced apoptosis. The IKK/NF-κB signaling pathway contributes to antiapoptotic gene expression. Pentoxifylline (PTX) inhibits IkB phosphorylation, blocking NF-κB and antiapoptotic activity. Methods We conducted a randomized, double-blind clinical trial on pediatric ALL patients undergoing induction therapy, assigning them to PTX or placebo group. Bone marrow aspirates were obtained on days 1, 8, 15, and 22. Apoptosis was assessed using Annexin-V/propidium iodide. Results Results indicated that the PTX group exhibited higher apoptosis on day-8 (41.3% vs. 19.4%, p =0.029) and day-15 (35.0% vs. 14.2%, p <0.01). On day-8, the PTX group displayed an MRD of 0.25% vs. 18.2% (p <0.01) in placebo group; on day-15, the PTX group demonstrated an MRD of 0.09% vs. 1.4% (p =0.02). Patients achieving an MRD <0.01% on day-8 demonstrated a 3-year Overall Survival (OS) of 81.6% vs. 58.3% (p =0.03); on day-15, patients with MRD <0.01% had a 3-year OS of 77.9% vs. 54.5% (p =0.03). The PTX group achieved an MRD of <0.01% earlier on days-8 and 15, along with a higher apoptosis rate, indicating a more favorable therapeutic response. In the entire cohort, patients achieving MRD <0.01% on day-8 or 15 displayed superior OS. Conclusion Our study demonstrates that PTX enhances apoptosis and reduces MRD in pediatric acute lymphoblastic leukemia patients. Clinical trial registration https://clinicaltrials.gov/, identifier NCT02451774.
Collapse
Affiliation(s)
- Violeta Salceda-Rivera
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Pablo C. Ortiz-Lazareno
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Georgina Hernández-Flores
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
| | - Jorge R. Vazquez-Urrutia
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Centro Universitario de Ciencias de la Salud, School of Medicine, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Jesus Meza-Arroyo
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Monzerrat Pardo-Zepeda
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Hugo Romo-Rubio
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Cesar Barba-Barba
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
| | - Fernando Sánchez-Zubieta
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
- Departamento de Clinicas de Reproduccion Humana, Crecimiento y Desarrollo Infantil, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Carlos Alfredo Barrón-Gallardo
- Departamento Académico de Disciplinas Especializantes de Ciencias de la Salud, Universidad Autonoma de Guadalajara, Zapopan, JAL, Mexico
| | - Oscar Gonzalez-Ramella
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Department of Pediatric Hemato-Oncology, Hospital Civil de Guadalajara “Dr. Juan I. Menchaca”, Guadalajara, JAL, Mexico
- Centro Universitario de Ciencias de la Salud, School of Medicine, Universidad de Guadalajara, Guadalajara, JAL, Mexico
| | - Alejandro Bravo-Cuellar
- Immunology Division, Western Biomedical Research Center, Mexican Social Security Institute, Guadalajara, JAL, Mexico
- Doctoral Program in Biomedical Sciences, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, JAL, Mexico
- Centro Universitario de los Altos, Universidad de Guadalajara, Tepatitlán de Morelos, JAL, Mexico
| |
Collapse
|
2
|
Sharew NT, Clark SR, Schubert KO, Amare AT. Pharmacogenomic scores in psychiatry: systematic review of current evidence. Transl Psychiatry 2024; 14:322. [PMID: 39107294 PMCID: PMC11303815 DOI: 10.1038/s41398-024-02998-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 08/10/2024] Open
Abstract
In the past two decades, significant progress has been made in the development of polygenic scores (PGSs). One specific application of PGSs is the development and potential use of pharmacogenomic- scores (PGx-scores) to identify patients who can benefit from a specific medication or are likely to experience side effects. This systematic review comprehensively evaluates published PGx-score studies in psychiatry and provides insights into their potential clinical use and avenues for future development. A systematic literature search was conducted across PubMed, EMBASE, and Web of Science databases until 22 August 2023. This review included fifty-three primary studies, of which the majority (69.8%) were conducted using samples of European ancestry. We found that over 90% of PGx-scores in psychiatry have been developed based on psychiatric and medical diagnoses or trait variants, rather than pharmacogenomic variants. Among these PGx-scores, the polygenic score for schizophrenia (PGSSCZ) has been most extensively studied in relation to its impact on treatment outcomes (32 publications). Twenty (62.5%) of these studies suggest that individuals with higher PGSSCZ have negative outcomes from psychotropic treatment - poorer treatment response, higher rates of treatment resistance, more antipsychotic-induced side effects, or more psychiatric hospitalizations, while the remaining studies did not find significant associations. Although PGx-scores alone accounted for at best 5.6% of the variance in treatment outcomes (in schizophrenia treatment resistance), together with clinical variables they explained up to 13.7% (in bipolar lithium response), suggesting that clinical translation might be achieved by including PGx-scores in multivariable models. In conclusion, our literature review found that there are still very few studies developing PGx-scores using pharmacogenomic variants. Research with larger and diverse populations is required to develop clinically relevant PGx-scores, using biology-informed and multi-phenotypic polygenic scoring approaches, as well as by integrating clinical variables with these scores to facilitate their translation to psychiatric practice.
Collapse
Affiliation(s)
- Nigussie T Sharew
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Asrat Woldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Division of Mental Health, Northern Adelaide Local Health Network, SA Health, Adelaide, Australia
- Headspace Adelaide Early Psychosis - Sonder, Adelaide, SA, Australia
| | - Azmeraw T Amare
- Discipline of Psychiatry, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
3
|
Saad AF, Pacheco LD, Saade GR. Immunosuppressant Medications in Pregnancy. Obstet Gynecol 2024; 143:e94-e106. [PMID: 38227938 DOI: 10.1097/aog.0000000000005512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/07/2023] [Indexed: 01/18/2024]
Abstract
Pregnant patients are often on immunosuppressant medications, most commonly to manage transplantation or autoimmune disorders. Most immunosuppressant agents, including tacrolimus, corticosteroids, azathioprine, and calcineurin inhibitors, are safe during pregnancy and lactation. However, mycophenolic acid is associated with higher risks of birth defects and should be avoided in pregnancy. Tacrolimus, the commonly used drug in transplantation medicine and autoimmune disorders, requires monitoring of serum levels for dose adjustment, particularly during pregnancy. Although no pregnancy-specific therapeutic range exists, the general target range is 5-15 ng/mL, and pregnant patients may require higher doses to achieve therapeutic levels. Adherence to prescribed immunosuppressive regimens is crucial to prevent graft rejection and autoimmune disorder flare-ups. This review aims to provide essential information about the use of immunosuppressant medications in pregnant individuals. With a rising number of pregnant patients undergoing organ transplantations or having autoimmune disorders, it is important to understand the implications of the use of these medications during pregnancy.
Collapse
Affiliation(s)
- Antonio F Saad
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Inova Fairfax, Fairfax, Virginia; the Division of Surgical Critical Care, Department of Anesthesiology, University of Texas Medical Branch at Galveston, Galveston, Texas; and the Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, Virginia
| | | | | |
Collapse
|
4
|
Nordengen AL, Kværner AS, Krutto A, Alavi DT, Henriksen HB, Henriksen C, Raastad T, Smeland S, Bøhn SK, Shaposhnikov S, Collins AR, Blomhoff R. DNA base oxidation in relation to TNM stages and chemotherapy treatment in colorectal cancer patients 2-9 months post-surgery. Free Radic Biol Med 2024; 212:174-185. [PMID: 38141887 DOI: 10.1016/j.freeradbiomed.2023.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/01/2023] [Accepted: 12/13/2023] [Indexed: 12/25/2023]
Abstract
Accumulation of DNA damage is a critical feature of genomic instability, which is a hallmark of various cancers. The enzyme-modified comet assay is a recognized method to detect specific DNA lesions at the level of individual cells. In this cross-sectional investigation, we explore possible links between clinicopathological and treatment related factors, nutritional status, physical activity and function, and DNA damage in a cohort of colorectal cancer (CRC) patients with non-metastatic disease. Levels of DNA damage in peripheral mononuclear blood cells (PBMCs) assessed 2-9 months post-surgery, were compared across tumour stage (localized (stage I-II) vs. regional (stage III) disease), localization (colon vs. rectosigmoid/rectum cancer), and adjuvant chemotherapy usage, with the last dosage administrated 2-191 days prior to sampling. Associations between DNA damage and indicators of nutritional status, physical activity and function were also explored. In PBMCs, DNA base oxidation was higher in patients diagnosed with regional compared with localized tumours (P = 0.03), but no difference was seen for DNA strand breaks (P > 0.05). Number of days since last chemotherapy dosage was negatively associated with DNA base oxidation (P < 0.01), and patients recently receiving chemotherapy (<15 days before blood collection) had higher levels of DNA base oxidation than those not receiving chemotherapy (P = 0.03). In the chemotherapy group, higher fat mass (in kg and %) as well as lower physical activity were associated with greater DNA base oxidation (P < 0.05). In conclusion, DNA base oxidation measured with the enzyme-modified comet assay varies according to tumour and lifestyle related factors in CRC patients treated for non-metastatic disease.
Collapse
Affiliation(s)
- Anne Lene Nordengen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway; Norgenotech AS, Oslo Cancer Cluster Incubator, Oslo, Norway; Department of Sport Science and Physical Education, Faculty of Health and Sport Sciences, University of Agder, Kristiansand, Norway.
| | - Ane S Kværner
- Section for Colorectal Cancer Screening, The Cancer Registry of Norway, Oslo, Norway
| | - Annika Krutto
- Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Dena T Alavi
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Hege B Henriksen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Christine Henriksen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Truls Raastad
- Department of Physical Performance, Norwegian School of Sport Science, Norway
| | - Sigbjørn Smeland
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Norway, Oslo, Norway
| | - Siv K Bøhn
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | | | | | - Rune Blomhoff
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Norway; Department of Clinical Service, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
5
|
Conroy MC, Lacey B, Bešević J, Omiyale W, Feng Q, Effingham M, Sellers J, Sheard S, Pancholi M, Gregory G, Busby J, Collins R, Allen NE. UK Biobank: a globally important resource for cancer research. Br J Cancer 2023; 128:519-527. [PMID: 36402876 PMCID: PMC9938115 DOI: 10.1038/s41416-022-02053-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/21/2022] Open
Abstract
UK Biobank is a large-scale prospective study with deep phenotyping and genomic data. Its open-access policy allows researchers worldwide, from academia or industry, to perform health research in the public interest. Between 2006 and 2010, the study recruited 502,000 adults aged 40-69 years from the general population of the United Kingdom. At enrolment, participants provided information on a wide range of factors, physical measurements were taken, and biological samples (blood, urine and saliva) were collected for long-term storage. Participants have now been followed up for over a decade with more than 52,000 incident cancer cases recorded. The study continues to be enhanced with repeat assessments, web-based questionnaires, multi-modal imaging, and conversion of the stored biological samples to genomic and other '-omic' data. The study has already demonstrated its value in enabling research into the determinants of cancer, and future planned enhancements will make the resource even more valuable to cancer researchers. Over 26,000 researchers worldwide are currently using the data, performing a wide range of cancer research. UK Biobank is uniquely placed to transform our understanding of the causes of cancer development and progression, and drive improvements in cancer treatment and prevention over the coming decades.
Collapse
Affiliation(s)
- Megan C Conroy
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK.
| | - Ben Lacey
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Jelena Bešević
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Wemimo Omiyale
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | - Qi Feng
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| | | | | | | | | | | | - John Busby
- UK Biobank, Stockport, Greater Manchester, UK
| | - Rory Collins
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
- UK Biobank, Stockport, Greater Manchester, UK
| | - Naomi E Allen
- Nuffield Department of Population Health (NDPH), University of Oxford, Oxford, UK
| |
Collapse
|
6
|
Günsel A, Yıldırım A, Taslimi P, Erden Y, Taskin-Tok T, Pişkin H, Bilgiçli AT, Gülçin İ, Nilüfer Yarasir M. Cytotoxicity effects and biochemical investigation of novel tetrakis-phthalocyanines bearing 2-thiocytosine moieties with molecular docking studies. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.109263] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
7
|
Saigusa H, Oyama A, Kitamura S, Asami H. Structural Characterization of 6-Thioguanosine and Its Monohydrate in the Gas Phase. J Phys Chem A 2021; 125:7217-7225. [PMID: 34433270 DOI: 10.1021/acs.jpca.1c05219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Detailed structural analysis of 6-thioguanosine (6TGs) in relation to its tautomerization and sugar conformation is performed in the gas phase using UV and IR spectroscopy combined with ab initio calculations. We have observed a thiol tautomer of 6TGs with its sugar moiety in the syn conformation that is stabilized by a strong intramolecular H-bonding between O5'H of the sugar and N3 atom of the guanine moiety. This observation is consistent with previous results for guanosine (Gs) in which the corresponding enol form is solely detected. We have also identified a monohydrate of 6TGs consisting of a thiol tautomer with the water linking guanine moiety and sugar OH group. It is demonstrated that hydration behavior of 6TGs is significantly different from that of Gs as a result of a weaker H-bonding ability of the thiol group.
Collapse
Affiliation(s)
- Hiroyuki Saigusa
- Graduate School for Bio- and Nanosystem Sciences, Yokohama City University, Yokohama 236-0027, Japan
| | - Ayumi Oyama
- Graduate School for Bio- and Nanosystem Sciences, Yokohama City University, Yokohama 236-0027, Japan
| | - Saki Kitamura
- Graduate School for Bio- and Nanosystem Sciences, Yokohama City University, Yokohama 236-0027, Japan
| | - Hiroya Asami
- Department of Chemistry, Faculty of Science, Gakushuin University, 1-5-1 Mejiro, Toshima-ku, Tokyo 171-8588, Japan
| |
Collapse
|
8
|
Boonyawat B, Monsereenusorn C, Photia A, Lertvivatpong N, Kaewchaivijit V, Jindatanmanusan P, Rujkijyanont P. ITPA:c.94C>A and NUDT15:c.415C>T Polymorphisms and Their Relation to Mercaptopurine-Related Myelotoxicity in Childhood Leukemia in Thailand. APPLICATION OF CLINICAL GENETICS 2021; 14:341-351. [PMID: 34349542 PMCID: PMC8326781 DOI: 10.2147/tacg.s318912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/19/2021] [Indexed: 11/30/2022]
Abstract
Background Mercaptopurine is a key agent in childhood leukemia treatment. Genetic polymorphism in the genes involving thiopurine metabolisms is related to 6-MP related toxicity. Objective This study aimed to determine the prevalence of ITPA:c.94C>A and NUDT15:c.415C>T polymorphisms among Thai children diagnosed with leukemia and their association with mercaptopurine-related myelotoxicity. Methods Patients and survivors with a diagnosis of leukemia treated with mercaptopurine-containing chemotherapy regimens were enrolled. Clinical data and laboratory parameters during treatment as well as ITPA:c.94C>A and NUDT15:c.415C>T genotypes were analyzed. Results In all, 99 patients with acute leukemia or survivors were enrolled in the study. The prevalences of ITPA:c.94C>A, NUDT15:c.415C>T, and co-occurrence of ITPA:c.94C>A and NUDT15:c.415C>T polymorphisms were 34, 17, and 4%, respectively. Numbers of absolute neutrophil count (ANC) and platelet count significantly decreased among patients carrying NUDT15:c.415C>T compared with NUDT15 wild type patients with p-values<0.001 and 0.019, respectively. The differences were not observed among patients carrying ITPA:c.94C>A compared with ITPA wild type patients. According to multivariate GEE, NUDT15:c.415C>T and co-occurrence of ITPA:c.94C>A and NUDT15:c.415C>T had a significant negative effect on ANC during treatment (coefficient: −463.81; CI: −778.53, −149.09; p-value=0.004 and coefficient: −527.56; CI: −1045.65, −9.48; p-value=0.046). No significant effect of ITPA:c.94C>A on ANC during treatment was observed. Conclusion ITPA:c.94C>A and NUDT15:c.415C>T polymorphisms are common among Thai children with leukemia. A strong association with mercaptopurine-related myelotoxicity was observed among patients carrying either NUDT15:c.415C>T alone or combined with ITPA:c.94C>A.
Collapse
Affiliation(s)
- Boonchai Boonyawat
- Division of Medical Genetics, Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Chalinee Monsereenusorn
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Apichat Photia
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Nawachai Lertvivatpong
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Varissara Kaewchaivijit
- Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Punyanuch Jindatanmanusan
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| | - Piya Rujkijyanont
- Division of Hematology-Oncology, Department of Pediatrics, Phramongkutklao College of Medicine and Phramongkutklao Hospital, Bangkok, Thailand
| |
Collapse
|
9
|
Vasistha A, Kothari R, Mishra A, De Andrés F, LLerena A, Nair S. Current Insights into Interethnic Variability in Testicular Cancers: Population Pharmacogenetics, Clinical Trials, Genetic Basis of Chemotherapy- Induced Toxicities and Molecular Signal Transduction. Curr Top Med Chem 2021; 20:1824-1838. [PMID: 32552648 DOI: 10.2174/1568026620666200618112205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/08/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022]
Abstract
Testicular cancer is an aggressive malignancy with a rising incidence rate across the globe. Testicular germ cell tumors are the most commonly diagnosed cancers, and surgical removal of the testes is often a radical necessity along with chemotherapy and radiotherapy. While seminomas are receptive to radiotherapy as well as chemotherapy, non-seminomatous germ cell tumors respond to chemotherapy only. Due to the singular nature of testicular cancers with associated orchiectomy and mortality, it is important to study the molecular basis and genetic underpinnings of this group of cancers across male populations globally. In this review, we shed light on the population pharmacogenetics of testicular cancer, pediatric and adult tumors, current clinical trials, genetic determinants of chemotherapy-induced toxicity in testicular cancer, as well as the molecular signal transduction pathways operating in this malignancy. Taken together, our discussions will help in enhancing our understanding of genetic factors in testicular carcinogenesis and chemotherapy-induced toxicity, augment our knowledge of this aggressive cancer at the cellular and molecular level, as well as improve precision medicine approaches to combat this disease.
Collapse
Affiliation(s)
- Aman Vasistha
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS University, V. L. Mehta Road, Vile Parle (West), Mumbai - 400 056, India
| | - Rishi Kothari
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, V. L. Mehta Road, Vile Parle (West), Mumbai - 400 056, India
| | - Adarsh Mishra
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's NMIMS University, V. L. Mehta Road, Vile Parle (West), Mumbai - 400 056, India
| | - Fernando De Andrés
- CICAB Clinical Research Centre at Extremadura University Hospital and Medical School, Universidad de Extremadura, Badajoz, Spain
| | - Adrián LLerena
- CICAB Clinical Research Centre at Extremadura University Hospital and Medical School, Universidad de Extremadura, Badajoz, Spain
| | - Sujit Nair
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, V. L. Mehta Road, Vile Parle (West), Mumbai - 400 056, India
| |
Collapse
|
10
|
Euceda N, Jahnke J, Espinal A, Louis MF, Bashkin E, Roccanova P, Espaillat A, Fuentes GV, Nieto F, Gao R. Thioguanine restoration through type I photosensitization-superoxide oxidation-glutathione reduction cycles. Phys Chem Chem Phys 2021; 23:5069-5073. [PMID: 33655288 DOI: 10.1039/d1cp00101a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
UVA-induced deleterious effect of thiopurine prodrugs including azathioprine, 6-mercaptopurine and 6-thioguanine (6-TG) increases the risk of cancer development due to the incorporation of 6-TG in patients' DNA. The catalytic mechanism by which thiobases act as a sustained oxidant producer has yet to be explored, especially through the Type I electron transfer pathway that produces superoxide radicals (O2˙-). Under Fenton-like conditions O2˙- radicals convert to extremely reactive hydroxyl radicals (˙OH), thus carrying even higher risk of biological damage than that induced by the well-studied type II reaction. By monitoring 6-TG/UVA-induced photochemistry in mass spectra and superoxide radicals (O2˙-) via nitro blue tetrazolium (NBT) reduction, this work provides two new findings: (1) in the presence of reduced glutathione (GSH), the production of O2˙-via the type I reaction is enhanced 10-fold. 6-TG thiyl radicals are identified as the primary intermediate formed in the reaction of 6-TG with O2˙-. The restoration of 6-TG and concurrent generation of O2˙- occur via a 3-step-cycle: 6-TG type I photosensitization, O2˙- oxidation and GSH reduction. (2) In the absence of GSH, 6-TG thiyl radicals undergo oxygen addition and sulfur dioxide removal to form carbon radicals (C6) which further convert to thioether by reacting with 6-TG molecules. These findings help explain not only thiol-regulation in a biological system but chemoprevention of cancer.
Collapse
Affiliation(s)
- Nelson Euceda
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - Joyce Jahnke
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - Aileen Espinal
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - Monique F Louis
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - Edan Bashkin
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - Patricia Roccanova
- Biological Sciences Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA
| | - Abraham Espaillat
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - German V Fuentes
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| | - Fernando Nieto
- Biological Sciences Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA
| | - Ruomei Gao
- Chemistry and Physics Department, SUNY College at Old Westbury, Old Westbury, NY 11568, USA.
| |
Collapse
|
11
|
Koukouli E, Wang D, Dondelinger F, Park J. A regularized functional regression model enabling transcriptome-wide dosage-dependent association study of cancer drug response. PLoS Comput Biol 2021; 17:e1008066. [PMID: 33493149 PMCID: PMC7920352 DOI: 10.1371/journal.pcbi.1008066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/01/2021] [Accepted: 12/17/2020] [Indexed: 11/18/2022] Open
Abstract
Cancer treatments can be highly toxic and frequently only a subset of the patient population will benefit from a given treatment. Tumour genetic makeup plays an important role in cancer drug sensitivity. We suspect that gene expression markers could be used as a decision aid for treatment selection or dosage tuning. Using in vitro cancer cell line dose-response and gene expression data from the Genomics of Drug Sensitivity in Cancer (GDSC) project, we build a dose-varying regression model. Unlike existing approaches, this allows us to estimate dosage-dependent associations with gene expression. We include the transcriptomic profiles as dose-invariant covariates into the regression model and assume that their effect varies smoothly over the dosage levels. A two-stage variable selection algorithm (variable screening followed by penalized regression) is used to identify genetic factors that are associated with drug response over the varying dosages. We evaluate the effectiveness of our method using simulation studies focusing on the choice of tuning parameters and cross-validation for predictive accuracy assessment. We further apply the model to data from five BRAF targeted compounds applied to different cancer cell lines under different dosage levels. We highlight the dosage-dependent dynamics of the associations between the selected genes and drug response, and we perform pathway enrichment analysis to show that the selected genes play an important role in pathways related to tumorigenesis and DNA damage response.
Collapse
Affiliation(s)
- Evanthia Koukouli
- Department of Mathematics and Statistics, Fylde College, Lancaster University, Bailrigg, Lancaster, UK
| | - Dennis Wang
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
- Department of Computer Science, University of Sheffield, Sheffield, UK
| | - Frank Dondelinger
- Centre for Health Informatics and Statistics, Lancaster Medical School, Lancaster University, Bailrigg, Lancaster, UK
| | - Juhyun Park
- Department of Mathematics and Statistics, Fylde College, Lancaster University, Bailrigg, Lancaster, UK
| |
Collapse
|
12
|
Chen X, Guo Y, Chen X. iGMDR: Integrated Pharmacogenetic Resource Guide to Cancer Therapy and Research. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:150-160. [PMID: 32916316 PMCID: PMC7646137 DOI: 10.1016/j.gpb.2019.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 09/27/2019] [Accepted: 11/29/2019] [Indexed: 10/25/2022]
Abstract
Current pharmacogenetic studies have obtained many genetic models that can predict the therapeutic efficacy of anticancer drugs. Although some of these models are of crucial importance and have been used in clinical practice, these very valuable models have not been well adopted into cancer research to promote the development of cancer therapies due to the lack of integration and standards for the existing data of the pharmacogenetic studies. For this purpose, we built a resource investigating genetic model of drug response (iGMDR), which integrates the models from in vitro and in vivo pharmacogenetic studies with different omics data from a variety of technical systems. In this study, we introduced a standardized process for all integrations, and described how users can utilize these models to gain insights into cancer. iGMDR is freely accessible at https://igmdr.modellab.cn.
Collapse
Affiliation(s)
- Xiang Chen
- Institute of Pharmaceutical Biotechnology and the First Affiliated Hospital Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Yi Guo
- Department of Polymer Science and Engineering and Key Laboratory of Adsorption and Separation Materials and Technologies of Zhejiang Province, Zhejiang University, Hangzhou 310027, China
| | - Xin Chen
- Institute of Pharmaceutical Biotechnology and the First Affiliated Hospital Department of Radiation Oncology, Zhejiang University School of Medicine, Hangzhou 310058, China; Joint Institute for Genetics and Genome Medicine between Zhejiang University and University of Toronto, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
13
|
Brady SC, Zdraljevic S, Bisaga KW, Tanny RE, Cook DE, Lee D, Wang Y, Andersen EC. A Novel Gene Underlies Bleomycin-Response Variation in Caenorhabditis elegans. Genetics 2019; 212:1453-1468. [PMID: 31171655 PMCID: PMC6707474 DOI: 10.1534/genetics.119.302286] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/30/2019] [Indexed: 12/14/2022] Open
Abstract
Bleomycin is a powerful chemotherapeutic drug used to treat a variety of cancers. However, individual patients vary in their responses to bleomycin. The identification of genetic differences that underlie this response variation could improve treatment outcomes by tailoring bleomycin dosages to each patient. We used the model organism Caenorhabditis elegans to identify genetic determinants of bleomycin-response differences by performing linkage mapping on recombinants derived from a cross between the laboratory strain (N2) and a wild strain (CB4856). This approach identified a small genomic region on chromosome V that underlies bleomycin-response variation. Using near-isogenic lines, and strains with CRISPR-Cas9 mediated deletions and allele replacements, we discovered that a novel nematode-specific gene (scb-1) is required for bleomycin resistance. Although the mechanism by which this gene causes variation in bleomycin responses is unknown, we suggest that a rare variant present in the CB4856 strain might cause differences in the potential stress-response function of scb-1 between the N2 and CB4856 strains, thereby leading to differences in bleomycin resistance.
Collapse
Affiliation(s)
- Shannon C Brady
- Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208
| | - Stefan Zdraljevic
- Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208
| | - Karol W Bisaga
- Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois 60208
| | - Robyn E Tanny
- Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
| | | | - Daehan Lee
- Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
| | - Ye Wang
- Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
| | - Erik C Andersen
- Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
- Interdisciplinary Biological Sciences Program, Northwestern University, Evanston, Illinois 60208
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
14
|
Di Mascio P, Martinez GR, Miyamoto S, Ronsein GE, Medeiros MHG, Cadet J. Singlet Molecular Oxygen Reactions with Nucleic Acids, Lipids, and Proteins. Chem Rev 2019; 119:2043-2086. [DOI: 10.1021/acs.chemrev.8b00554] [Citation(s) in RCA: 253] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Paolo Di Mascio
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, CP 26077, CEP 05508-000, São Paulo, SP Brazil
| | - Glaucia R. Martinez
- Departamento de Bioquímica e Biologia Molecular, Setor de Ciências Biológicas, Universidade Federal do Paraná, 81531-990 Curitiba, PR, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, CP 26077, CEP 05508-000, São Paulo, SP Brazil
| | - Graziella E. Ronsein
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, CP 26077, CEP 05508-000, São Paulo, SP Brazil
| | - Marisa H. G. Medeiros
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, CP 26077, CEP 05508-000, São Paulo, SP Brazil
| | - Jean Cadet
- Département de Médecine Nucléaire et Radiobiologie, Faculté de Médecine des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, J1H 5N4 Québec, Canada
| |
Collapse
|
15
|
Xia Y, Wang F, Wang RN, Liu KH, Su HM. Reaction kinetics between thiobases and singlet oxygen studied by direct detection of the 1O2 luminescence decay. CHINESE J CHEM PHYS 2019. [DOI: 10.1063/1674-0068/cjcp1811268] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Ye Xia
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Fei Wang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ren-nian Wang
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Kun-hui Liu
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hong-mei Su
- College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
16
|
Palmirotta R, Carella C, Silvestris E, Cives M, Stucci SL, Tucci M, Lovero D, Silvestris F. SNPs in predicting clinical efficacy and toxicity of chemotherapy: walking through the quicksand. Oncotarget 2018; 9:25355-25382. [PMID: 29861877 PMCID: PMC5982750 DOI: 10.18632/oncotarget.25256] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/07/2018] [Indexed: 12/19/2022] Open
Abstract
In the "precision medicine" era, chemotherapy still remains the backbone for the treatment of many cancers, but no affordable predictors of response to the chemodrugs are available in clinical practice. Single nucleotide polymorphisms (SNPs) are gene sequence variations occurring in more than 1% of the full population, and account for approximately 80% of inter-individual genomic heterogeneity. A number of studies have investigated the predictive role of SNPs of genes enrolled in both pharmacodynamics and pharmacokinetics of chemotherapeutics, but the clinical implementation of related results has been modest so far. Among the examined germline polymorphic variants, several SNPs of dihydropyrimidine dehydrogenase (DPYD) and uridine diphosphate glucuronosyltransferases (UGT) have shown a robust role as predictors of toxicity following fluoropyrimidine- and/or irinotecan-based treatments respectively, and a few guidelines are mandatory in their detection before therapy initiation. Contrasting results, however, have been reported on the capability of variants of other genes as MTHFR, TYMS, ERCC1, XRCC1, GSTP1, CYP3A4/3A5 and ABCB1, in predicting either therapy efficacy or toxicity in patients undergoing treatment with pyrimidine antimetabolites, platinum derivatives, irinotecan and taxanes. While formal recommendations for routine testing of these SNPs cannot be drawn at this moment, therapeutic decisions may indeed benefit of germline genomic information, when available. Here, we summarize the clinical impact of germline genomic variants on the efficacy and toxicity of major chemodrugs, with the aim to facilitate the therapeutic expectance of clinicians in the odiern quicksand field of complex molecular biology concepts and controversial trial data interpretation.
Collapse
Affiliation(s)
- Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Claudia Carella
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Erica Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Mauro Cives
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Stefania Luigia Stucci
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Marco Tucci
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Domenica Lovero
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, Section of Clinical and Molecular Oncology, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
17
|
Kværner AS, Minaguchi J, Yamani NE, Henriksen C, Ræder H, Paur I, Henriksen HB, Wiedswang G, Smeland S, Blomhoff R, Collins AR, Bøhn SK. DNA damage in blood cells in relation to chemotherapy and nutritional status in colorectal cancer patients—A pilot study. DNA Repair (Amst) 2018; 63:16-24. [DOI: 10.1016/j.dnarep.2018.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/10/2018] [Accepted: 01/10/2018] [Indexed: 02/08/2023]
|
18
|
Yeh CM, Su SC, Lin CW, Yang WE, Chien MH, Reiter RJ, Yang SF. Melatonin as a potential inhibitory agent in head and neck cancer. Oncotarget 2017; 8:90545-90556. [PMID: 29163852 PMCID: PMC5685773 DOI: 10.18632/oncotarget.20079] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/26/2017] [Indexed: 12/29/2022] Open
Abstract
Melatonin is a molecule secreted by the pineal gland; it is an important regulator of sleep and circadian rhythms. Through multiple interrelated mechanisms, melatonin exhibits various inhibitory properties at different stages of tumor progression. Many studies have explored the oncostatic effects of melatonin on hormone-dependent tumors. In this review, we highlight recent advances in understanding the effects of melatonin on the development of head and neck cancers, including molecular mechanisms identified through experimental and clinical observations. Because melatonin exerts a wide range of effects, melatonin may influence many mechanisms that influence the development of cancer. These include cell proliferation, apoptosis, angiogenesis, extracellular matrix remodeling through matrix metalloproteinases, and genetic polymorphism. Thus, the evidence discussed in this article will serve as a basis for basic and clinical research to promote the use of melatonin for understanding and controlling the development of head and neck cancers.
Collapse
Affiliation(s)
- Chia-Ming Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shih-Chi Su
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Russel J Reiter
- Department of Cellular and Structural Biology, The University of Texas Health Science Center, San Antonio, TX, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
19
|
Finkelstein Y, Blonquist TM, Vijayanathan V, Stevenson KE, Neuberg DS, Silverman LB, Vrooman LM, Sallan SE, Cole PD. A thymidylate synthase polymorphism is associated with increased risk for bone toxicity among children treated for acute lymphoblastic leukemia. Pediatr Blood Cancer 2017; 64. [PMID: 27957785 DOI: 10.1002/pbc.26393] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/03/2016] [Accepted: 11/15/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Bone fractures and osteonecrosis frequently complicate therapy for childhood acute lymphoblastic leukemia (ALL). Bone toxicity has been associated with exposure to corticosteroids and methotrexate (MTX) and age greater than 10 years. We tested whether common genetic polymorphisms were associated with bone toxicity during treatment for ALL. PROCEDURE A total of 615 of 794 children enrolled on Dana Farber Cancer Institute ALL Consortium protocol 05-001 (NCT00400946) met eligibility criteria for inclusion in this analysis. Nineteen candidate polymorphisms were selected a priori, targeting genes related to glucocorticoid metabolism, oxidative damage, and folate physiology. Polymorphisms were genotyped using either PCR-based allelic discrimination or PCR product length analysis. RESULTS Twenty percent of subjects were homozygous for two 28 bp repeats (2R/2R, where 2R is two 28-nucleotide repeats within the 5' untranslated region [UTR] of the thymidylate synthase [TS] gene) within the 5' UTR of the gene for TS. This 2R/2R genotype was associated with increased risk of osteonecrosis among children younger than 10 years at diagnosis (multivariable hazard ratio [HR] 2.71; 95% confidence interval [CI] 1.23-5.95; P = 0.013), and with bone fracture among children ≥ 10 years (multivariable HR 2.10; 95% CI 1.11-3.96; P = 0.022). No significant association was observed between TS genotype and red blood cell (RBC) folate, RBC MTX, or relapse risk. CONCLUSIONS A common genetic variant is associated with increased risk of osteonecrosis among children younger than 10 years at diagnosis and with bone fractures among older children. These findings suggest that children and adolescents with the 2R/2R TS genotype should be closely monitored for the development of bone toxicity during therapy for ALL, and support a clinical trial testing the efficacy of protective interventions specifically in this vulnerable population.
Collapse
Affiliation(s)
| | | | - Veena Vijayanathan
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Lewis B Silverman
- Dana Farber Cancer Institute, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Lynda M Vrooman
- Dana Farber Cancer Institute, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Stephen E Sallan
- Dana Farber Cancer Institute, Boston, Massachusetts.,Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Peter D Cole
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
20
|
Arslancan S, Martínez-Fernández L, Corral I. Photophysics and Photochemistry of Canonical Nucleobases’ Thioanalogs: From Quantum Mechanical Studies to Time Resolved Experiments. Molecules 2017. [PMCID: PMC6152766 DOI: 10.3390/molecules22060998] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Interest in understanding the photophysics and photochemistry of thiated nucleobases has been awakened because of their possible involvement in primordial RNA or their potential use as photosensitizers in medicinal chemistry. The interpretation of the photodynamics of these systems, conditioned by their intricate potential energy surfaces, requires the powerful interplay between experimental measurements and state of the art molecular simulations. In this review, we provide an overview on the photophysics of natural nucleobases’ thioanalogs, which covers the last 30 years and both experimental and computational contributions. For all the canonical nucleobase’s thioanalogs, we have compiled the main steady state absorption and emission features and their interpretation in terms of theoretical calculations. Then, we revise the main topographical features, including stationary points and interstate crossings, of their potential energy surfaces based on quantum mechanical calculations and we conclude, by combining the outcome of different spectroscopic techniques and molecular dynamics simulations, with the mechanism by which these nucleobase analogs populate their triplet excited states, which are at the origin of their photosensitizing properties.
Collapse
Affiliation(s)
- Serra Arslancan
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain;
| | - Lara Martínez-Fernández
- Istituto Biostrutture e Bioimmagini-Consiglio Nazionale delle Ricerche, Via Mezzocannone 16, Napoli I-80134, Italy
- Correspondence: (L.M.-F.); (I.C.); Tel.: +34-91-497-8471 (I.C.)
| | - Inés Corral
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Madrid 28049, Spain;
- Institute for Advanced Research in Chemical Sciences (IADCHEM), Universidad Autónoma de Madrid, Madrid 28049, Spain
- Correspondence: (L.M.-F.); (I.C.); Tel.: +34-91-497-8471 (I.C.)
| |
Collapse
|
21
|
Lievano F, Scarazzini L, Shen F, Duhig J, Jokinen J. The future of safety science is happening now: The modernization of the benefit-risk paradigm. Pharmacoepidemiol Drug Saf 2017; 26:869-874. [DOI: 10.1002/pds.4241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/28/2017] [Accepted: 05/07/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Fabio Lievano
- Pharmacovigilance and Patient Safety; AbbVie Inc.; North Chicago IL USA
| | - Linda Scarazzini
- Pharmacovigilance and Patient Safety; AbbVie Inc.; North Chicago IL USA
| | - Frank Shen
- Pharmacovigilance and Patient Safety; AbbVie Inc.; North Chicago IL USA
| | - James Duhig
- Pharmacovigilance and Patient Safety; AbbVie Inc.; North Chicago IL USA
| | - Jeremy Jokinen
- Pharmacovigilance and Patient Safety; AbbVie Inc.; North Chicago IL USA
| |
Collapse
|
22
|
Ankathil R. ABCB1 genetic variants in leukemias: current insights into treatment outcomes. Pharmgenomics Pers Med 2017; 10:169-181. [PMID: 28546766 PMCID: PMC5438075 DOI: 10.2147/pgpm.s105208] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Despite improvements in treatment of different types of leukemia, not all patients respond optimally for a particular treatment. Some treatments will work better for some, while being harmful or ineffective for others. This is due to genetic variation in the form of single-nucleotide polymorphisms (SNPs) that affect gene expression or function and cause inherited interindividual differences in the metabolism and disposition of drugs. Drug transporters are one of the determinants governing the pharmacokinetic profile of chemotherapeutic drugs. The ABCB1 transporter gene transports a wide range of drugs, including drugs used in leukemia treatment. Polymorphisms in the ABCB1 gene do affect intrinsic resistance and pharmacokinetics of several drugs used in leukemia treatment protocols and thereby affect the efficacy of treatment and event-free survival. This review focuses on the impact of three commonly occurring SNPs (1236C>T, 2677G>T/A, and 3435C>T) of ABCB1 on treatment response of various types of leukemia. From the literature available, some of the genotypes and haplotypes of these SNPs have been found to be potential determinants of interindividual variability in drug disposition and pharmacologic response in different types of leukemia. However, due to inconsistencies in the results observed across the studies, additional studies, considering novel genomic methodologies, comprehensive definition of clinical phenotypes, adequate sample size, and uniformity in all the confounding factors, are warranted.
Collapse
Affiliation(s)
- Ravindran Ankathil
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
23
|
Biological and predictive role of ERCC1 polymorphisms in cancer. Crit Rev Oncol Hematol 2017; 111:133-143. [PMID: 28259288 DOI: 10.1016/j.critrevonc.2017.01.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/14/2017] [Accepted: 01/24/2017] [Indexed: 12/22/2022] Open
Abstract
Excision repair cross-complementation group 1 (ERCC1) is a key component in DNA repair mechanisms and may influence the tumor DNA-targeting effect of the chemotherapeutic agent oxaliplatin. Germline ERCC1 polymorphisms may alter the protein expression and published data on their predictive and prognostic value have so far been contradictory. In the present article we review available evidence on the clinical role and utility of ERCC1 polymorphisms and, in the absence of a 'perfect' trial, what we call the 'sliding doors' trial, we present the data of ERCC1 genotyping in our local patient population. We found a useful predictive value for oxaliplatin-induced risk of anemia.
Collapse
|
24
|
Abstract
The nutrient folate is essential for normal cell growth and development. Enzymes that require folate as a cofactor have been common targets for chemotherapeutic agents. Limited clinical guidance is available regarding optimal levels of dietary or supplemental folate intake during antifolate chemotherapy to meet the goals of minimizing treatment-related toxicity yet preserving treatment effectiveness. Patients with higher folate intake may experience less treatment-related toxicities; however, the concern is that folate may interfere with the effectiveness of the antifolate treatment and possibly support tumor growth. This article will review folate and antifolate metabolism and review the current body of knowledge regarding the interactions between folates and antifolates. Future research in this area should include evaluation of pretreatment folate status and dietary and supplemental folate intake before, during, and after treatment.
Collapse
Affiliation(s)
- Kim Robien
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, PO Box 19024, M4-B402 Seattle, Washington 98109-1024, USA.
| |
Collapse
|
25
|
Burgis NE. A disease spectrum for ITPA variation: advances in biochemical and clinical research. J Biomed Sci 2016; 23:73. [PMID: 27770805 PMCID: PMC5075207 DOI: 10.1186/s12929-016-0291-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/19/2016] [Indexed: 12/15/2022] Open
Abstract
Human ITPase (encoded by the ITPA gene) is a protective enzyme which acts to exclude noncanonical (deoxy)nucleoside triphosphates ((d)NTPs) such as (deoxy)inosine 5′-triphosphate ((d)ITP), from (d)NTP pools. Until the last few years, the importance of ITPase in human health and disease has been enigmatic. In 2009, an article was published demonstrating that ITPase deficiency in mice is lethal. All homozygous null offspring died before weaning as a result of cardiomyopathy due to a defect in the maintenance of quality ATP pools. More recently, a whole exome sequencing project revealed that very rare, severe human ITPA mutation results in early infantile encephalopathy and death. It has been estimated that nearly one third of the human population has an ITPA status which is associated with decreased ITPase activity. ITPA status has been linked to altered outcomes for patients undergoing thiopurine or ribavirin therapy. Thiopurine therapy can be toxic for patients with ITPA polymorphism, however, ITPA polymorphism is associated with improved outcomes for patients undergoing ribavirin treatment. ITPA polymorphism has also been linked to early-onset tuberculosis susceptibility. These data suggest a spectrum of ITPA-related disease exists in human populations. Potentially, ITPA status may affect a large number of patient outcomes, suggesting that modulation of ITPase activity is an important emerging avenue for reducing the number of negative outcomes for ITPA-related disease. Recent biochemical studies have aimed to provide rationale for clinical observations, better understand substrate selectivity and provide a platform for modulation of ITPase activity.
Collapse
Affiliation(s)
- Nicholas E Burgis
- Department of Chemistry and Biochemistry, Eastern Washington University, 226 Science Building, Cheney, WA, 99004, USA.
| |
Collapse
|
26
|
Fakhoury M, Jacqz-Aigrain E, de Beaumais T, Médard Y. [Not Available]. Therapie 2016; 65:187-93. [PMID: 27392985 DOI: 10.2515/therapie/2010031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Accepted: 03/23/2010] [Indexed: 12/25/2022]
Abstract
6-mercaptopurine, a key drug for the treatment of acute lymphoblastic leukaemia in children, is a prodrug metabolized into 6-thioguanine (6-TGN) which are the active compounds and into methylated metabolites, primary by thiopurine S-methyltransferase enzyme (TPMT). This enzyme displays important inter subject variability linked to a genetic polymorphism: when treated with standard doses of thiopurine, TPMT-deficient and heterozygous patients are at great risk for developing severe and potentially life-threatening toxicity (hematopoietic, hepatic, mucositis. . . ) but show a better survival rate while patients with high TPMT activity (wild type) present lower peripheral red blood cells 6-TGN concentrations and a higher risk of leukemia relapse. Genotyping remains crucial before 6-MP administration at diagnosis to identify patients with homozygous mutant TPMT genotype and therefore prevent severe and life-threatening toxicity, and to individualize therapy according to TMPT genotype. Follow-up of ALL treatment should preferentially be based on repeated determinations of intracellular active metabolites (6-thioguanine nucleotides) and methylated metabolites in addition to haematological surveillance.
Collapse
Affiliation(s)
- May Fakhoury
- Laboratoire de Pharmacologie Pédiatrique et Pharmacogénétique, Hôpital Robert Debré, Paris, France
| | - Evelyne Jacqz-Aigrain
- Laboratoire de Pharmacologie Pédiatrique et Pharmacogénétique, Hôpital Robert Debré, Paris, France.
| | | | | | | |
Collapse
|
27
|
Zhao Y, Liang M, Li X, Fan K, Xiao J, Li Y, Shi H, Wang F, Choi HS, Cheng D, Yan X. Bioengineered Magnetoferritin Nanoprobes for Single-Dose Nuclear-Magnetic Resonance Tumor Imaging. ACS NANO 2016; 10:4184-4191. [PMID: 26959856 DOI: 10.1021/acsnano.5b07408] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Despite all the advances in multimodal imaging, it remains a significant challenge to acquire both magnetic resonance and nuclear imaging in a single dose because of the enormous difference in sensitivity. Indeed, nuclear imaging is almost 10(6)-fold more sensitive than magnetic resonance imaging (MRI); thus, repeated injections are generally required to obtain sufficient MR signals after nuclear imaging. Here, we show that strategically engineered magnetoferritin nanoprobes can image tumors with high sensitivity and specificity using SPECT and MRI in living mice after a single intravenous injection. The magnetoferritin nanoprobes composed of (125)I radionuclide-conjugated human H-ferritin iron nanocages ((125)I-M-HFn) internalize robustly into cancer cells via a novel tumor-specific HFn-TfR1 pathway. In particular, the endocytic recycling characteristic of TfR1 transporters solves the nuclear signal blocking issue caused by the high dose nanoprobes injected for MRI, thus enabling simultaneous functional and morphological tumor imaging without reliance on multi-injections.
Collapse
Affiliation(s)
- Yanzhao Zhao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University/Shanghai Institute of Medical Imaging , Shanghai 200032, China
| | - Minmin Liang
- Key Laboratory of Protein and Peptide Pharmaceutical/Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology/Beijing Translational Engineering Center of Biomacromolecular Drugs, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Xiao Li
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University/Shanghai Institute of Medical Imaging , Shanghai 200032, China
| | - Kelong Fan
- Key Laboratory of Protein and Peptide Pharmaceutical/Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology/Beijing Translational Engineering Center of Biomacromolecular Drugs, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Jie Xiao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University/Shanghai Institute of Medical Imaging , Shanghai 200032, China
| | - Yanli Li
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University/Shanghai Institute of Medical Imaging , Shanghai 200032, China
| | - Hongcheng Shi
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University/Shanghai Institute of Medical Imaging , Shanghai 200032, China
| | - Fei Wang
- Key Laboratory of Protein and Peptide Pharmaceutical/Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology/Beijing Translational Engineering Center of Biomacromolecular Drugs, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| | - Hak Soo Choi
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School , Boston, Massachusetts 02215, United States
| | - Dengfeng Cheng
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University/Shanghai Institute of Medical Imaging , Shanghai 200032, China
| | - Xiyun Yan
- Key Laboratory of Protein and Peptide Pharmaceutical/Chinese Academy of Sciences-University of Tokyo Joint Laboratory of Structural Virology and Immunology/Beijing Translational Engineering Center of Biomacromolecular Drugs, Institute of Biophysics, Chinese Academy of Sciences , Beijing 100101, China
| |
Collapse
|
28
|
Singh HO, Lata S, Angadi M, Bapat S, Pawar J, Nema V, Ghate MV, Sahay S, Gangakhedkar RR. Impact of GSTM1, GSTT1 and GSTP1 gene polymorphism and risk of ARV-associated hepatotoxicity in HIV-infected individuals and its modulation. THE PHARMACOGENOMICS JOURNAL 2015; 17:53-60. [PMID: 26667829 DOI: 10.1038/tpj.2015.88] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/15/2015] [Accepted: 11/02/2015] [Indexed: 12/29/2022]
Abstract
Glutathione S-transferase (GST) family is involved in a two-stage detoxification process of a wide range of environmental toxins, carcinogen and antiretroviral (ARV) therapy (ART) drugs. The aim of this study is to describe the impact of genetic polymorphisms of GSTM1, GSTT1 and GSTP1-313A/G in the risk of ARV-associated hepatotoxicity in HIV-infected individuals and its modulation in hepatotoxic patients. We enrolled a total of 34 patients with hepatotoxicity, 131 HIV-infected individuals without hepatotoxicity under non-nucleoside reverse transcriptase inhibitor containing ART and 153 unrelated healthy individuals. With a case-control design, polymorphisms of GSTM1, GSTT1 and GSTP1-313A/G gene were genotyped by PCR and restriction enzyme-length polymorphism. Genotypes of GSTT1 null were significantly higher in HIV-infected individuals as compared with healthy controls (P=0.01, odds ratio (OR)=1.54). HIV-infected individuals with GSTM1-null genotype showed higher risk (P=0.09, OR=1.37) for hepatotoxicity, but risk was not significant. On evaluating gene-gene interaction models, GSTM1 null and GSTT1 null showed significant association with the risk of hepatotoxicity in HIV-infected individuals (P=0.004, OR=2.67) owing to synergistic effect of these genes. Individuals with GSTT1-null and GSTM1-null genotypes showed higher risk of hepatotoxicity with advanced stage of (CD4<200) of HIV infection (P=0.18, OR=1.39; P=0.63, OR=1.13). In case-only analysis, GSTT1-null genotype among alcohol users showed elevated risk of hepatotoxicity in HIV-infected individuals (P=0.12, OR=1.36, 95% confidence interval (CI): 0.94-1.97) as compared with GSTT1 genotypes. The carriers GSTM1-null+GSTT1-null genotype among nevirapine user showed prominent risk of hepatotoxicity in HIV-infected individuals (P=0.12, OR=4.21, 95% CI: 0.60-29.54). Hence, we can conclude that GSTT1-null and GSTM1-null genotypes alone and in combination may predict the acquisition of hepatotoxicity.
Collapse
Affiliation(s)
- H O Singh
- Department of Molecular Biology, National AIDS Research Institute, Pune, India
| | - S Lata
- Department of Molecular Biology, National AIDS Research Institute, Pune, India
| | - M Angadi
- Department of Clinical Sciences, National AIDS Research Institute, Pune, India
| | - S Bapat
- Department of Clinical Sciences, National AIDS Research Institute, Pune, India
| | - J Pawar
- Department of Clinical Sciences, National AIDS Research Institute, Pune, India
| | - V Nema
- Department of Molecular Biology, National AIDS Research Institute, Pune, India
| | - M V Ghate
- Department of Clinical Sciences, National AIDS Research Institute, Pune, India
| | - S Sahay
- Department of Clinical Sciences, National AIDS Research Institute, Pune, India
| | - R R Gangakhedkar
- Department of Clinical Sciences, National AIDS Research Institute, Pune, India
| |
Collapse
|
29
|
Johnatty SE, Tyrer JP, Kar S, Beesley J, Lu Y, Gao B, Fasching PA, Hein A, Ekici AB, Beckmann MW, Lambrechts D, Van Nieuwenhuysen E, Vergote I, Lambrechts S, Rossing MA, Doherty JA, Chang-Claude J, Modugno F, Ness RB, Moysich KB, Levine DA, Kiemeney LA, Massuger LFAG, Gronwald J, Lubiński J, Jakubowska A, Cybulski C, Brinton L, Lissowska J, Wentzensen N, Song H, Rhenius V, Campbell I, Eccles D, Sieh W, Whittemore AS, McGuire V, Rothstein JH, Sutphen R, Anton-Culver H, Ziogas A, Gayther SA, Gentry-Maharaj A, Menon U, Ramus SJ, Pearce CL, Pike MC, Stram DO, Wu AH, Kupryjanczyk J, Dansonka-Mieszkowska A, Rzepecka IK, Spiewankiewicz B, Goodman MT, Wilkens LR, Carney ME, Thompson PJ, Heitz F, du Bois A, Schwaab I, Harter P, Pisterer J, Hillemanns P, Karlan BY, Walsh C, Lester J, Orsulic S, Winham SJ, Earp M, Larson MC, Fogarty ZC, Høgdall E, Jensen A, Kjaer SK, Fridley BL, Cunningham JM, Vierkant RA, Schildkraut JM, Iversen ES, Terry KL, Cramer DW, Bandera EV, Orlow I, Pejovic T, Bean Y, Høgdall C, Lundvall L, McNeish I, Paul J, Carty K, Siddiqui N, Glasspool R, Sellers T, Kennedy C, Chiew YE, Berchuck A, MacGregor S, Pharoah PDP, Goode EL, deFazio A, Webb PM, Chenevix-Trench G. Genome-wide Analysis Identifies Novel Loci Associated with Ovarian Cancer Outcomes: Findings from the Ovarian Cancer Association Consortium. Clin Cancer Res 2015; 21:5264-76. [PMID: 26152742 PMCID: PMC4624261 DOI: 10.1158/1078-0432.ccr-15-0632] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/20/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Chemotherapy resistance remains a major challenge in the treatment of ovarian cancer. We hypothesize that germline polymorphisms might be associated with clinical outcome. EXPERIMENTAL DESIGN We analyzed approximately 2.8 million genotyped and imputed SNPs from the iCOGS experiment for progression-free survival (PFS) and overall survival (OS) in 2,901 European epithelial ovarian cancer (EOC) patients who underwent first-line treatment of cytoreductive surgery and chemotherapy regardless of regimen, and in a subset of 1,098 patients treated with ≥ 4 cycles of paclitaxel and carboplatin at standard doses. We evaluated the top SNPs in 4,434 EOC patients, including patients from The Cancer Genome Atlas. In addition, we conducted pathway analysis of all intragenic SNPs and tested their association with PFS and OS using gene set enrichment analysis. RESULTS Five SNPs were significantly associated (P ≤ 1.0 × 10(-5)) with poorer outcomes in at least one of the four analyses, three of which, rs4910232 (11p15.3), rs2549714 (16q23), and rs6674079 (1q22), were located in long noncoding RNAs (lncRNAs) RP11-179A10.1, RP11-314O13.1, and RP11-284F21.8, respectively (P ≤ 7.1 × 10(-6)). ENCODE ChIP-seq data at 1q22 for normal ovary show evidence of histone modification around RP11-284F21.8, and rs6674079 is perfectly correlated with another SNP within the super-enhancer MEF2D, expression levels of which were reportedly associated with prognosis in another solid tumor. YAP1- and WWTR1 (TAZ)-stimulated gene expression and high-density lipoprotein (HDL)-mediated lipid transport pathways were associated with PFS and OS, respectively, in the cohort who had standard chemotherapy (pGSEA ≤ 6 × 10(-3)). CONCLUSIONS We have identified SNPs in three lncRNAs that might be important targets for novel EOC therapies.
Collapse
Affiliation(s)
- Sharon E Johnatty
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jonathan P Tyrer
- Department of Oncology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Siddhartha Kar
- Department of Oncology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Jonathan Beesley
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Yi Lu
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Bo Gao
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia. Center for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Peter A Fasching
- Division of Hematology and Oncology, Department of Medicine, University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, California. University Hospital Erlangen, Institute of Human Genetics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Alexander Hein
- University Hospital Erlangen, Department of Gynecology and Obstetrics, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Arif B Ekici
- University Hospital Erlangen, Institute of Human Genetics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias W Beckmann
- University Hospital Erlangen, Department of Gynecology and Obstetrics, Friedrich-Alexander-University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Diether Lambrechts
- Vesalius Research Center, VIB, Leuven, Belgium. Laboratory for Translational Genetics, Department of Oncology, University of Leuven, Leuven, Belgium
| | - Els Van Nieuwenhuysen
- Department of Gynecologic Oncology, Leuven Cancer Institute, University of Leuven, Leuven, Belgium
| | - Ignace Vergote
- Department of Gynecologic Oncology, Leuven Cancer Institute, University of Leuven, Leuven, Belgium
| | - Sandrina Lambrechts
- Department of Gynecologic Oncology, Leuven Cancer Institute, University of Leuven, Leuven, Belgium
| | - Mary Anne Rossing
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, University of Washington, Seattle, Washington
| | - Jennifer A Doherty
- Department of Community and Family Medicine, Section of Biostatistics and Epidemiology, The Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Jenny Chang-Claude
- German Cancer Research Center (DKFZ), Division of Cancer Epidemiology, Heidelberg, Germany
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania. Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania. Women's Cancer Research Program, Magee-Women's Research Institute and University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Roberta B Ness
- The University of Texas School of Public Health, Houston, Texas
| | - Kirsten B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - Douglas A Levine
- Gynecology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Lambertus A Kiemeney
- Radboud University Medical Centre, Radboud Institute for Health Sciences, Nijmegen, the Netherlands
| | - Leon F A G Massuger
- Radboud University Medical Centre, Radboud Institute for Molecular Sciences, Nijmegen, the Netherlands
| | - Jacek Gronwald
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubiński
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Anna Jakubowska
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Cezary Cybulski
- International Hereditary Cancer Center, Department of Genetics and Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Louise Brinton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Nicolas Wentzensen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Honglin Song
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Valerie Rhenius
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Ian Campbell
- Cancer Genetics Laboratory, Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Diana Eccles
- Faculty of Medicine, University of Southampton, University Hospital Southampton, Southampton, Hampshire, United Kingdom
| | - Weiva Sieh
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, California
| | - Alice S Whittemore
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, California
| | - Valerie McGuire
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, California
| | - Joseph H Rothstein
- Department of Health Research and Policy - Epidemiology, Stanford University School of Medicine, Stanford, California
| | - Rebecca Sutphen
- Epidemiology Center, College of Medicine, University of South Florida, Tampa, Florida
| | - Hoda Anton-Culver
- Department of Epidemiology, Center for Cancer Genetics Research and Prevention, School of Medicine, University of California Irvine, Irvine, California
| | - Argyrios Ziogas
- Department of Epidemiology, University of California Irvine, Irvine, California
| | - Simon A Gayther
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | | | - Usha Menon
- Women's Cancer, UCL EGA Institute for Women's Health, London, United Kingdom
| | - Susan J Ramus
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | - Celeste L Pearce
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California. Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan
| | - Malcolm C Pike
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California. Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Daniel O Stram
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | - Anna H Wu
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | - Jolanta Kupryjanczyk
- Department of Pathology and Laboratory Diagnostics, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Agnieszka Dansonka-Mieszkowska
- Department of Pathology and Laboratory Diagnostics, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Iwona K Rzepecka
- Department of Pathology and Laboratory Diagnostics, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Beata Spiewankiewicz
- Department of Gynecologic Oncology, The Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Marc T Goodman
- Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California. Community and Population Health Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Michael E Carney
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Pamela J Thompson
- Cancer Prevention and Control, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California. Community and Population Health Research Institute, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany. Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany. Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | - Ira Schwaab
- Institut für Humangenetik Wiesbaden, Wiesbaden, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany. Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
| | | | - Peter Hillemanns
- Department of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Beth Y Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Christine Walsh
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sandra Orsulic
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Stacey J Winham
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Madalene Earp
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Melissa C Larson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Zachary C Fogarty
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Estrid Høgdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark. Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Susanne Kruger Kjaer
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Copenhagen, Denmark. Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Brooke L Fridley
- Biostatistics and Informatics Shared Resource, University of Kansas Medical Center, Kansas City, Kansas
| | - Julie M Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Robert A Vierkant
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Joellen M Schildkraut
- Department of Community and Family Medicine, Duke University Medical Center, Durham, North Carolina. Cancer Control and Population Sciences, Duke Cancer Institute, Durham, North Carolina
| | - Edwin S Iversen
- Department of Statistical Science, Duke University, Durham, North Carolina
| | - Kathryn L Terry
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Harvard School of Public Health, Boston, Massachusetts
| | - Daniel W Cramer
- Obstetrics and Gynecology Epidemiology Center, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts. Harvard School of Public Health, Boston, Massachusetts
| | - Elisa V Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute of New Jersey, The State University of New Jersey, New Brunswick, New Jersey
| | - Irene Orlow
- Memorial Sloan Kettering Cancer Center, Department of Epidemiology and Biostatistics, Epidemiology Service, New York, New York
| | - Tanja Pejovic
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon. Knight Cancer Institute, Portland, Oregon
| | - Yukie Bean
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, Oregon. Knight Cancer Institute, Portland, Oregon
| | - Claus Høgdall
- Department of Gynaecology, The Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lene Lundvall
- Department of Gynaecology, The Juliane Marie Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ian McNeish
- Institute of Cancer Sciences, University of Glasgow, Wolfson Wohl Cancer Research Centre, Beatson Institute for Cancer Research, Glasgow, United Kingdom
| | - James Paul
- Cancer Research UK Clinical Trials Unit, Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Karen Carty
- Cancer Research UK Clinical Trials Unit, Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Nadeem Siddiqui
- Department of Gynaecological Oncology, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Rosalind Glasspool
- Cancer Research UK Clinical Trials Unit, Glasgow, The Beatson West of Scotland Cancer Centre, Glasgow, United Kingdom
| | - Thomas Sellers
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Catherine Kennedy
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia. Center for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Yoke-Eng Chiew
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia. Center for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina
| | - Stuart MacGregor
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Paul D P Pharoah
- Department of Oncology, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge, United Kingdom
| | - Ellen L Goode
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Anna deFazio
- Department of Gynaecological Oncology, Westmead Hospital, Sydney, New South Wales, Australia. Center for Cancer Research, University of Sydney at Westmead Millennium Institute, Sydney, New South Wales, Australia
| | - Penelope M Webb
- Department of Population Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Georgia Chenevix-Trench
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
30
|
Kodidela S, Pradhan SC, Dubashi B, Basu D. Influence of dihydrofolate reductase gene polymorphisms rs408626 (-317A>G) and rs442767 (-680C>A) on the outcome of methotrexate-based maintenance therapy in South Indian patients with acute lymphoblastic leukemia. Eur J Clin Pharmacol 2015; 71:1349-58. [PMID: 26335211 DOI: 10.1007/s00228-015-1930-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023]
Abstract
PURPOSE The most common cause of treatment failure in acute lymphoblastic leukaemia (ALL) is the relapse. Genetic polymorphisms of dihydrofolate reductase (DHFR) enzyme affect the response to methotrexate (MTX) treatment. Inter-individual variability exists in the distribution of DHFR variants, and they influence MTX treatment outcome. To the best of our knowledge, there are no genetic studies reported from India, which have explored the influence of DHFR variants on the outcome of MTX treatment. Therefore, we aim to study the influence of DHFR rs408626 (-317A>G) and rs442767 (-680C>A) variants on ALL outcome in South Indian patients. METHODS A total of 70 ALL patients who were on MTX-based maintenance therapy were recruited for the study. DNA was extracted from leukocytes, and genotyping was done by real-time PCR. RESULTS The DHFR-317GG genotype was associated with the increased risk of relapse in patients with ALL (relative risk 2.25, 95% confidence interval (CI) 1.38 to 3.6, p = 0.02). DHFR-317AA and -680CA genotypes were found to be associated with severe leucopenia (p < 0.05). In Cox regression model, -317GG genotype was found to have lower relapse-free survival (hazard ratio (HR) 2.56, 95% CI 1.06 to 6.19, p = 0.03) and overall survival (HR 3.72, 95% CI 1.44 to 9.65, p = 0.007). Similarly, patients with white blood cell (WBC) count >50,000 cells/mm(3) were also found to have lower relapse-free survival (HR 2.20, 95% CI 1.10 to 4.79, p = 0.04) and overall survival (HR 3.30, 95% CI 1.45 to 7.53, p = 0.004). CONCLUSION The GG genotype of DHFR-317A>G variant is associated with increased risk of ALL relapse and lower overall survival in South Indian population. Both variants of DHFR (-317 AA and -680 CA) are found to be associated with severe leucopenia caused by MTX.
Collapse
Affiliation(s)
- Sunitha Kodidela
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Gorimedu, Puducherry, India.
| | - Suresh Chandra Pradhan
- Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Gorimedu, Puducherry, India
| | - Biswajit Dubashi
- Department of Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Gorimedu, Puducherry, India
| | - Debdatta Basu
- Department of Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Gorimedu, Puducherry, India
| |
Collapse
|
31
|
McNeer JL, Raetz EA. Childhood Acute Lymphoblastic Leukemia: Toward Personalized Medicine. CURRENT PEDIATRICS REPORTS 2015. [DOI: 10.1007/s40124-015-0078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
32
|
Keung YK, Keung LW, Hong-Lung Hu E. A case of recurrent pancytopenia in a patient with acute promyelocytic leukemia on maintenance chemotherapy and concomitant methyltetrahydrofolate reductase and thiopurine S-methyltransferase mutation - review of literature. J Oncol Pharm Pract 2015; 22:548-51. [DOI: 10.1177/1078155215577235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Pharmacogenetics is a study of how genetic variation of an individual affects the drug response. We report a case of recurrent pancytopenia resulting from maintenance chemotherapy in a patient with acute promyelocytic leukemia and two pharmacogenetic mutations, namely, methylene tetrahydrofolate reductase C677T homozygous mutation and thiopurine methyltransferase mutation.
Collapse
Affiliation(s)
- Yi-Kong Keung
- UCLA, Hematology-Oncology Division Clinic, Alhambra, USA
| | - Lap-Woon Keung
- UCLA, Hematology-Oncology Division Clinic, Alhambra, USA
| | | |
Collapse
|
33
|
Zhang S, Tan IB, Sapari NS, Grabsch HI, Okines A, Smyth EC, Aoyama T, Hewitt LC, Inam I, Bottomley D, Nankivell M, Stenning SP, Cunningham D, Wotherspoon A, Tsuburaya A, Yoshikawa T, Soong R, Tan P. Technical reproducibility of single-nucleotide and size-based DNA biomarker assessment using DNA extracted from formalin-fixed, paraffin-embedded tissues. J Mol Diagn 2015; 17:242-50. [PMID: 25746798 DOI: 10.1016/j.jmoldx.2014.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 12/02/2014] [Accepted: 12/05/2014] [Indexed: 02/07/2023] Open
Abstract
DNA extracted from formalin-fixed, paraffin-embedded (FFPE) tissues has been used in the past to analyze genetic polymorphisms. We evaluated the technical reproducibility of different types of assays for gene polymorphisms using DNA extracted from FFPE material. By using the MassARRAY iPLEX system, we investigated polymorphisms in DPYD (rs1801159 and rs3918290), UMPS (rs1801019), ERCC1 (rs11615), ERCC1 (rs3212986), and ERCC2 (rs13181) in 56 FFPE DNA samples. By using PCR, followed by size-based gel electrophoresis, we also examined TYMS 5' untranslated region 2R/3R repeats and GSTT1 deletions in 50 FFPE DNA samples and 34 DNAs extracted from fresh-frozen tissues and cell lines. Each polymorphism was analyzed by two independent runs. We found that iPLEX biomarker assays measuring single-nucleotide polymorphisms provided consistent concordant results. However, by using FFPE DNA, size-based PCR biomarkers (GSTT1 and TYMS 5' untranslated region) were discrepant in 32.7% (16/49, with exact 95% CI, 19.9%-47.5%; exact binomial confidence limit test) and 4.2% (2/48, with exact 95% CI, 0.5%-14.3%) of cases, respectively, whereas no discrepancies were observed using intact genomic DNA. Our findings suggest that DNA from FFPE material can be used to reliably test single-nucleotide polymorphisms. However, results based on size-based PCR biomarkers, and particularly GSTT1 deletions, using FFPE DNA need to be interpreted with caution. Independent repeated assays should be performed on all cases to assess potential discrepancies.
Collapse
Affiliation(s)
- Shenli Zhang
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Iain B Tan
- Department of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Nur S Sapari
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Heike I Grabsch
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Alicia Okines
- Department of Gastrointestinal Oncology, Royal Marsden Hospital, Sutton, United Kingdom
| | - Elizabeth C Smyth
- Department of Gastrointestinal Oncology, Royal Marsden Hospital, Sutton, United Kingdom
| | - Toru Aoyama
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Lindsay C Hewitt
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Imran Inam
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Dan Bottomley
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Matthew Nankivell
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Sally P Stenning
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - David Cunningham
- Department of Gastrointestinal Oncology, Royal Marsden Hospital, Sutton, United Kingdom
| | | | - Akira Tsuburaya
- Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Department of Pathology, National University Health System, Singapore, Singapore
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Graduate Medical School, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Department of Cellular and Molecular Research, National Cancer Centre, Singapore, Singapore; Cancer Therapeutics and Stratified Oncology Group, Genome Institute of Singapore, Singapore, Singapore.
| |
Collapse
|
34
|
Rizzi V, Losito I, Ventrella A, Fini P, Fraix A, Sortino S, Agostiano A, Longobardi F, Cosma P. Rose Bengal-photosensitized oxidation of 4-thiothymidine in aqueous medium: evidence for the reaction of the nucleoside with singlet state oxygen. Phys Chem Chem Phys 2015; 17:26307-19. [DOI: 10.1039/c5cp03615a] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Thymidine and a thiothymidine dimer are the products from the reaction of 4-thiothymidine with 1O2 generated through Rose Bengal-mediated photosensitization.
Collapse
Affiliation(s)
- Vito Rizzi
- Università degli Studi “Aldo Moro” di Bari
- Dip. Chimica
- 4-70126 Bari
- Italy
| | - Ilario Losito
- Università degli Studi “Aldo Moro” di Bari
- Dip. Chimica
- 4-70126 Bari
- Italy
- Centro Interdipartimentale SMART
| | - Andrea Ventrella
- Università degli Studi “Aldo Moro” di Bari
- Dip. Chimica
- 4-70126 Bari
- Italy
| | - Paola Fini
- Consiglio Nazionale delle Ricerche CNR-IPCF
- UOS Bari
- 4-70126 Bari
- Italy
| | - Aurore Fraix
- Laboratory of Photochemistry
- Department of Drug Sciences
- University of Catania
- I-95125 Catania
- Italy
| | - Salvatore Sortino
- Laboratory of Photochemistry
- Department of Drug Sciences
- University of Catania
- I-95125 Catania
- Italy
| | - Angela Agostiano
- Università degli Studi “Aldo Moro” di Bari
- Dip. Chimica
- 4-70126 Bari
- Italy
- Consiglio Nazionale delle Ricerche CNR-IPCF
| | | | - Pinalysa Cosma
- Università degli Studi “Aldo Moro” di Bari
- Dip. Chimica
- 4-70126 Bari
- Italy
- Consiglio Nazionale delle Ricerche CNR-IPCF
| |
Collapse
|
35
|
Aráoz HV, D’Aloi K, Foncuberta ME, Sanchez La Rosa CG, Alonso CN, Chertkoff L, Felice M. Pharmacogenetic studies in children with acute lymphoblastic leukemia in Argentina. Leuk Lymphoma 2014; 56:1370-8. [DOI: 10.3109/10428194.2014.951844] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
36
|
Rizzi V, Losito I, Ventrella A, Fini P, Agostiano A, Longobardi F, Cosma P. pH-related features and photostability of 4-thiothymidine in aqueous solution: an investigation by UV-visible, NMR and FTIR-ATR spectroscopies and by electrospray ionization mass spectrometry. RSC Adv 2014. [DOI: 10.1039/c4ra09747e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
37
|
Drum M, Kranaster R, Ewald C, Blasczyk R, Marx A. Variants of a Thermus aquaticus DNA polymerase with increased selectivity for applications in allele- and methylation-specific amplification. PLoS One 2014; 9:e96640. [PMID: 24800860 PMCID: PMC4011760 DOI: 10.1371/journal.pone.0096640] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/09/2014] [Indexed: 01/25/2023] Open
Abstract
The selectivity of DNA polymerases is crucial for many applications. For example, high discrimination between the extension of matched versus mismatched primer termini is desired for the detection of a single nucleotide variation at a particular locus within the genome. Here we describe the generation of thermostable mutants of the large fragment of Thermus aquaticus DNA polymerase (KlenTaq) with increased mismatch extension selectivity. In contrast to previously reported much less active KlenTaq mutants with mismatch discrimination abilities, many of the herein discovered mutants show conserved wild-type-like high activities. We demonstrate for one mutant containing the single amino acid exchange R660V the suitability for application in allele-specific amplifications directly from whole blood without prior sample purification. Also the suitability of the mutant for methylation specific amplification in the diagnostics of 5-methyl cytosines is demonstrated. Furthermore, the identified mutant supersedes other commercially available enzymes in human leukocyte antigen (HLA) analysis by sequence-specific primed polymerase chain reactions (PCRs).
Collapse
Affiliation(s)
- Matthias Drum
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Ramon Kranaster
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
- myPOLS Biotec, University of Konstanz, Konstanz, Germany
| | - Christina Ewald
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Andreas Marx
- Department of Chemistry, Konstanz Research School Chemical Biology, University of Konstanz, Konstanz, Germany
- * E-mail:
| |
Collapse
|
38
|
Gillis NK, Patel JN, Innocenti F. Clinical implementation of germ line cancer pharmacogenetic variants during the next-generation sequencing era. Clin Pharmacol Ther 2014; 95:269-80. [PMID: 24136381 PMCID: PMC4128332 DOI: 10.1038/clpt.2013.214] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/12/2013] [Indexed: 01/13/2023]
Abstract
More than 100 medications approved by the US Food and Drug Administration include pharmacogenetic biomarkers in the drug label, many with cancer indications referencing germ line DNA variations. With the advent of next-generation sequencing (NGS) and its rapidly increasing uptake into cancer research and clinical practice, an enormous amount of data to inform documented gene-drug associations will be collected that must be exploited to optimize patient benefit. This review focuses on the implementation of germ line cancer pharmacogenetics in clinical practice. Specifically, it discusses the importance of germ line variation in cancer and the role of NGS in pharmacogenetic discovery and implementation. In the context of a scenario in which massive amounts of NGS-based genetic information will be increasingly available to health stakeholders, this review explores the ongoing debate regarding the threshold of evidence necessary for implementation, provides an overview of recommendations in cancer by professional organizations and regulatory bodies, and discusses limitations of current guidelines and strategies to improve third-party coverage.
Collapse
Affiliation(s)
- Nancy K. Gillis
- Eshelman School of Pharmacy, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC
| | - Jai N. Patel
- Eshelman School of Pharmacy, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC
| | - Federico Innocenti
- Eshelman School of Pharmacy, Institute for Pharmacogenomics and Individualized Therapy, University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
39
|
Yang JJ, Bhojwani D. Thiopurine S-methyltransferase pharmacogenetics in childhood acute lymphoblastic leukemia. Methods Mol Biol 2014; 999:273-84. [PMID: 23666706 DOI: 10.1007/978-1-62703-357-2_20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
Pharmacogenetics is the growing field of study of genetic variations underlying interindividual differences in drug response. Inherited polymorphisms in genes coding for drug-metabolizing enzymes, transporters, and targets influence toxicity as well as efficacy associated with the medication. Thiopurines are agents widely used in hematologic malignancies, transplantation, and chronic inflammatory conditions. Myelosuppression is the commonly encountered dose-limiting toxicity. Polymorphisms in the thiopurine S-methyltransferase gene (TPMT), the predominant inactivating enzyme for thiopurines in hematopoietic tissue, are correlated with enzymatic activity of TPMT, thiopurine metabolism, and risk of clinical toxicity. In this chapter, we present TPMT genotype assessment that allows for prescribing pharmacogenetically guided doses to enhance patient safety and drug efficacy.
Collapse
Affiliation(s)
- Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
40
|
Genetic variations in stem cell-related genes and colorectal cancer prognosis. J Gastrointest Cancer 2013; 43:584-93. [PMID: 22528324 DOI: 10.1007/s12029-012-9388-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Many properties of cancer cells are reminiscent of those in normal stem cells. Genes important to stem cell development have been significantly implicated in the etiology and clinical outcome of colorectal cancer (CRC). However, the associations of genetic variations in these genes with CRC prognosis have not yet been elucidated. METHODS We analyzed the effects of eight potentially functional single nucleotide polymorphisms (SNPs) in six stem cell-related genes on the prognosis of a well-characterized population of 380 Chinese CRC patients diagnosed from February 2006 to January 2010. RESULTS The most significant finding was related to rs879882, a variant in the 5' region of POU5F1 gene which encodes a protein essential for embryonic stem cell self-renewal and pluripotency, and induced pluripotent stem cell reprogramming. The variant-containing genotypes of rs879882 were associated with an increased risk of recurrence (hazard ratio [HR] = 2.10, 95% confidence interval [CI] 1.17-3.76, P = 0.01). In chemotherapy-stratified analysis, the association remained borderline significant in patients receiving chemotherapy (HR = 1.97, 95% CI 0.89-4.34, P = 0.09). In addition, a nonsynonymous SNP of APC gene was also significantly associated with recurrence risk in chemotherapy-treated patients (HR = 2.63, 95% CI 1.14-6.06 P = 0.02). Further analyses showed a combined effect of the two SNPs in predicting CRC recurrence in patients receiving chemotherapy (P = 0.04) but not in those without chemotherapy (P = 0.43). Moreover, an exploratory multivariate assessment model indicated that these two variants enhanced the power to predict recurrence after chemotherapy. CONCLUSION We presented one of the first epidemiologic studies showing that stem cell-related genetic variants may impact CRC clinical outcomes, especially in chemotherapy-treated patients.
Collapse
|
41
|
Armenian SH, Landier W, Hudson MM, Robison LL, Bhatia S. Children's Oncology Group's 2013 blueprint for research: survivorship and outcomes. Pediatr Blood Cancer 2013; 60:1063-8. [PMID: 23255494 PMCID: PMC3799776 DOI: 10.1002/pbc.24422] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 11/07/2012] [Indexed: 12/12/2022]
Abstract
Improvements in the treatment of childhood cancer have resulted in over 360,000 survivors of childhood cancer in the U.S. There is now a heightened recognition of the need to reduce treatment-related sequelae and optimize the quality of life of children treated for cancer. Survivorship studies conducted in the cooperative group setting have provided us with important information on long-term intellectual function, organ toxicity, reproductive outcomes, second cancers, late mortality, and disparities in outcomes. Ongoing health education initiatives have helped standardize the follow-up care for childhood cancer survivors and facilitate the early transfer of health-related information to patients, families, and healthcare providers.
Collapse
Affiliation(s)
| | - Wendy Landier
- Department of Population Sciences, City of Hope, Duarte, CA
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Leslie L. Robison
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN
| | - Smita Bhatia
- Department of Population Sciences, City of Hope, Duarte, CA
| | | |
Collapse
|
42
|
Wu H, Xie J, Pan Q, Wang B, Hu D, Hu X. Anticancer agent shikonin is an incompetent inducer of cancer drug resistance. PLoS One 2013; 8:e52706. [PMID: 23300986 PMCID: PMC3536779 DOI: 10.1371/journal.pone.0052706] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 11/19/2012] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Cancer drug resistance is a major obstacle for the success of chemotherapy. Since most clinical anticancer drugs could induce drug resistance, it is desired to develop candidate drugs that are highly efficacious but incompetent to induce drug resistance. Numerous previous studies have proven that shikonin and its analogs not only are highly tumoricidal but also can bypass drug-transporter and apoptotic defect mediated drug resistance. The purpose of this study is to investigate if or not shikonin is a weak inducer of cancer drug resistance. EXPERIMENTAL DESIGN Different cell lines (K562, MCF-7, and a MDR cell line K562/Adr), after repeatedly treated with shikonin for 18 months, were assayed for drug resistance and gene expression profiling. RESULTS After 18-month treatment, cells only developed a mere 2-fold resistance to shikonin and a marginal resistance to cisplatin and paclitaxel, without cross resistance to shikonin analogs and other anticancer agents. Gene expression profiles demonstrated that cancer cells did strongly respond to shikonin treatment but failed to effectively mobilize drug resistant machineries. Shikonin-induced weak resistance was associated with the up-regulation of βII-tubulin, which physically interacted with shikonin. CONCLUSION Taken together, apart from potent anticancer activity, shikonin and its analogs are weak inducers of cancer drug resistance and can circumvent cancer drug resistance. These merits make shikonin and its analogs potential candidates for cancer therapy with advantages of avoiding induction of drug resistance and bypassing existing drug resistance.
Collapse
Affiliation(s)
- Hao Wu
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiansheng Xie
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiangrong Pan
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Beibei Wang
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Danqing Hu
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xun Hu
- Cancer Institute, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
43
|
Relling MV. Pharmacogenomics of Adverse Effects of Anti-Leukemic Agents in Children. J Pediatr Pharmacol Ther 2012; 17:7-11. [DOI: 10.5863/1551-6776-17.1.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Mary V Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, and the University of Tennessee Health Science Center, College of Pharmacy, Memphis, Tennessee
| |
Collapse
|
44
|
Koomdee N, Hongeng S, Apibal S, Pakakasama S. Association Between Polymorphisms of Dihydrofolate Reductase and Gamma Glutamyl Hydrolase Genes and Toxicity of High Dose Methotrexate in Children with Acute Lymphoblastic Leukemia. Asian Pac J Cancer Prev 2012; 13:3461-4. [DOI: 10.7314/apjcp.2012.13.7.3461] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
45
|
Sepe DM, McWilliams T, Chen J, Kershenbaum A, Zhao H, La M, Devidas M, Lange B, Rebbeck TR, Aplenc R. Germline genetic variation and treatment response on CCG-1891. Pediatr Blood Cancer 2012; 58:695-700. [PMID: 21618417 PMCID: PMC3165089 DOI: 10.1002/pbc.23192] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 04/12/2011] [Indexed: 01/30/2023]
Abstract
BACKGROUND Recent studies suggest that polymorphisms in genes encoding enzymes involved in drug detoxification and metabolism may influence disease outcome in pediatric acute lymphoblastic leukemia (ALL). We sought to extend current knowledge by using standard and novel statistical methodology to examine polymorphic variants of genes and relapse risk, toxicity, and drug dose delivery in standard risk ALL. PROCEDURE We genotyped and abstracted chemotherapy drug dose data from treatment roadmaps on 557 patients on the Children's Cancer Group ALL study, CCG-1891. Fourteen common polymorphisms in genes involved in folate metabolism and/or phase I and II drug detoxification were evaluated individually and clique-finding methodology was employed for detection of significant gene-gene interactions. RESULTS After controlling for known risk factors, polymorphisms in four genes: GSTP1*B (HR = 1.94, P = 0.047), MTHFR (HR = 1.61, P = 0.034), MTRR (HR = 1.95, P = 0.01), and TS (3R/4R, HR = 3.69, P = 0.007) were found to significantly increase relapse risk. One gene-gene pair, MTRR A/G and GSTM1 null genotype, significantly increased the risk of relapse after correction for multiple comparisons (P = 0.012). Multiple polymorphisms were associated with various toxicities and there was no significant difference in dose of chemotherapy delivered by genotypes. CONCLUSIONS These data suggest that various polymorphisms play a role in relapse risk and toxicity during childhood ALL therapy and that genotype does not play a role in adjustment of drug dose administered. Additionally, gene-gene interactions may increase the risk of relapse in childhood ALL and the clique method may have utility in further exploring these interactions. childhood ALL therapy.
Collapse
Affiliation(s)
- Dana M. Sepe
- The Children’s Hospital of Philadelphia, Philadelphia, PA, University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Jinbo Chen
- University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Huaqing Zhao
- The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Mei La
- Children’s Oncology Group, Arcadia, CA
| | - Meenakshi Devidas
- Children’s Oncology Group Statistics and Data Center and University of Florida College of Medicine, Gainesville, FL
| | - Beverly Lange
- The Children’s Hospital of Philadelphia, Philadelphia, PA, University of Pennsylvania School of Medicine, Philadelphia, PA
| | | | - Richard Aplenc
- The Children’s Hospital of Philadelphia, Philadelphia, PA, University of Pennsylvania School of Medicine, Philadelphia, PA
| |
Collapse
|
46
|
Hofbauer GFL, Attard NR, Harwood CA, McGregor JM, Dziunycz P, Iotzova-Weiss G, Straub G, Meyer R, Kamenisch Y, Berneburg M, French LE, Wüthrich RP, Karran P, Serra AL. Reversal of UVA skin photosensitivity and DNA damage in kidney transplant recipients by replacing azathioprine. Am J Transplant 2012; 12:218-25. [PMID: 21943390 DOI: 10.1111/j.1600-6143.2011.03751.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Azathioprine is associated with enhanced skin photosensitivity to ultraviolet A (UVA) and leads to incorporation of 6-thioguanine (6-TG) into DNA of dividing cells. Unlike canonical DNA, 6-TG DNA is damaged by UVA, which comprises more than 90% of the ultraviolet reaching earth. Skin photosensitivity to UVA and UVB was measured in 48 kidney transplant patients immunosuppressed either by azathioprine (n = 32) or mycophenolate (n = 16). In 23 patients, azathioprine was subsequently replaced by mycophenolate and skin photosensitivity, DNA 6-TG content in peripheral blood mononuclear cells, and susceptibility to UVA-induced DNA damage were monitored for up to 2 years. The mean minimal erythema dose to UVA on azathioprine was twofold lower than on mycophenolate. Three months after replacing azathioprine by mycophenolate mofetil, the minimal erythema dose to UVA had increased from 15 to 25 J/cm(2) (p < 0.001) accompanied by reduced DNA 6-TG content. P53 protein expression in irradiated skin indicated reduced susceptibility to UVA-induced DNA damage. 6-TG DNA in peripheral blood mononuclear cells remained measurable for over 2 years. Replacing azathioprine selectively reduced the skin photosensitivity to UVA, attenuated UVA-induced skin DNA damage, and is likely based on incorporated 6-TG in DNA.
Collapse
Affiliation(s)
- G F L Hofbauer
- Department of Dermatology, University Hospital Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Thiopurines were examined for their ability to produce singlet oxygen ((1)O(2)) with UVA light. The target compounds were three thiopurine prodrugs, azathioprine (Aza), 6-mercaptopurine (6-MP) and 6-thioguanine (6-TG), and their S-methylated derivatives of 6-methylmercaptopurine (me6-MP) and 6-methylthioguanine (me6-TG). Our results showed that these thiopurines were efficient (1)O(2) sensitizers under UVA irradiation but rapidly lost their photoactivities for (1)O(2) production over time by a self-sensitized photooxidation of sulfur atoms in the presence of oxygen and UVA light. The initial quantum yields of (1)O(2) production were determined to be in the range of 0.30-0.6 in aqueous solutions. Substitution of a hydrogen atom with a nitroimidazole or methyl group at S decreased the efficacy of photosensitized (1)O(2) production as found for Aza, me6-MP and me6-TG. (1)O(2)-induced formation of 8-oxo-7,8-dihydro-2'-dexyguanosine (8-oxodGuo) was assessed by incubation of 6-methylthiopurine/UVA-treated calf thymus DNA with human repair enzyme 8-oxodGuo DNA glycosylase (hOGG1), followed by apurinic (AP) site determination. Because more 8-oxodGuo was formed in Tris D(2)O than in Tris H(2)O, (1)O(2) is implicated as a key species in the reaction. These findings provided quantitative information on the photosensitization efficacy of thiopurines and to some extent revealed the correlations between photoactivity and phototoxicity.
Collapse
|
48
|
Kasaian K, Jones SJ. A new frontier in personalized cancer therapy: mapping molecular changes. Future Oncol 2011; 7:873-94. [PMID: 21732758 DOI: 10.2217/fon.11.63] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in the genome of a normal cell can affect the function of its many genes and pathways. These alterations could eventually transform the cell from a normal to a malignant state by allowing an uncontrolled proliferation of the cell and formation of a cancer tumor. Each tumor in an individual patient can have hundreds of mutated genes and perturbed pathways. Cancers clinically presenting as the same type or subtype could potentially be very different at the molecular level and thus behave differently in response to therapy. The challenge is to distinguish the key mutations driving the cancer from the background of mutational noise and find ways to effectively target them. The promise is that such a molecular approach to classifying cancer will lead to better diagnostic, prognostic and personalized treatment strategies. This article provides an overview of advances in the molecular characterization of cancers and their applications in therapy.
Collapse
Affiliation(s)
- Katayoon Kasaian
- Canada's Michael Smith Genome Sciences Centre, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | |
Collapse
|
49
|
Gutmann DH, Stiles CD, Lowe SW, Bollag GE, Furnari FB, Charest AL. Report from the fifth National Cancer Institute Mouse Models of Human Cancers Consortium Nervous System Tumors Workshop. Neuro Oncol 2011; 13:692-9. [PMID: 21727208 DOI: 10.1093/neuonc/nor080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Cancers of the nervous system are clinically challenging tumors that present with varied histopathologies and genetic etiologies. While the prognosis for the most malignant of these tumors is essentially unchanged despite decades of basic and translational science research, the past few years have witnessed the identification of numerous targetable molecular alterations in these cancers. With the advent of advanced genomic sequencing methodologies and the development of accurate small-animal models of these nervous system cancers, we are now ideally positioned to develop personalized therapies that target the unique cellular and molecular changes that define their formation and drive their continued growth. Recently, the National Cancer Institute convened a workshop to advance our understanding of nervous system cancer mouse models and to inform clinical trials by reconsidering these neoplasms as complex biological systems characterized by heterogeneity at all levels.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | |
Collapse
|
50
|
Polymorphism of the thymidylate synthase gene and risk of relapse in childhood ALL. Leuk Res 2011; 35:1464-6. [PMID: 21550658 DOI: 10.1016/j.leukres.2011.04.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 03/21/2011] [Accepted: 04/07/2011] [Indexed: 11/24/2022]
Abstract
Polymorphisms of genes encoding proteins involved in drug metabolism can influence the efficacy of leukemia treatment. In this population-wide study we aimed to evaluate selected, metabolically active genetic polymorphisms as prognostic markers of treatment efficacy in acute lymphoblastic leukemia (ALL). A total of 51 cases of leukemia relapse were diagnosed in a group of 354 patients with ALL. A strong association between promoter tandem repeat polymorphism of the thymidylate synthase gene and the relapse frequency was found. We believe that genotyping for this variant should be performed in patients treated for ALL to enable further optimizing of treatment protocols.
Collapse
|