Supplementary Table 1. Summary of FDA-Approved Anticancer Cytotoxic Drugs at May 2019

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Table 1. Summary of FDA-Approved Anticancer Cytotoxic Drugs at May 2019 Supplementary table 1. Summary of FDA-approved anticancer cytotoxic drugs at May 2019. Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References Substrate of Overexpression and lower CYP3A4, phosphorylation of CDC2, enhanced DNA Busulfan CML GSTA1, GSTA2, Unclear [1–3] stress response, inactivating mutations in GSTM1, GSTP1, the mismatch repair system (in vitro) MGST2. Alkylsulfonate DNA Altretamine Ovarian Unclear Unclear Unclear [3] Anal, bladder, breast, cervical, gastric, lip, mesothelioma, Substrate of DNA Mitomycin Unclear Overexpression of ABC transporters [3,4] NSCLC, oral cavity, POR pancreas, peritoneum, and Ethyleneimine pharynx Breast, ovarian, Substrate of Inhibitor of BCHE, DNA Thiotepa Enhanced activity of GST [3,5] bladder CYP3A4 CYP2B6 Synthetic drugs Synthetic Alkylating Agents Alkylating DNA, HL, Substrate of Inactivating mutations in the mismatch MAOA, Procarbazine oligodendrogliomas, Unclear [6,7] CYP1B1, XDH repair system (in vitro) MAOB PCNSL Methylhydrazine Substrate of DNA Bendamustine CLL, NHL Unclear Overexpression of CD69 (in vitro) [8] CYP1A2 Nitrogen mustard derivative mustard Nitrogen SUPPLEMENTARY TABLE 1. SUMMARY OF FDA-APPROVED ANTICANCER CYTOTOXIC DRUGS AT MAY 2019 (CONTINUATION) Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References CLL, lymphomas (HL, NHL, giant Substrate of Overexpression of GST isoenzymes and DNA Chlorambucil Unclear [3,9] folicular and GSTP1 ABCC1 lymphosarcoma) ALL, brain, breast, leukemias, malignant Increased levels of glutathione and increased DNA Cyclophosphamide lymphomas, Unclear NR1I2 [3,10,11] activity of GST and ALDH1 multiple myeloma, ovarian, retinoblastoma Substrate of CYP2A6, CYP2B6, CYP2C8, NR1I2; inducer of TP53 silencing, increased GST and ALDH1 Lymphomas, CYP2C9, CYP2C8, inhibitor activities, increased DNA repair, DNA Ifosfamide sarcomas, [3,12] CYP2C18, and inducer of overexpression of thioredoxin reductase (in testicular CYP2C19, CYP3A4 vitro) CYP3A4, CYP3A5, Synthetic drugs Synthetic Alkylating Agents Alkylating PTGS1 Nitrogen mustard derivative mustard Nitrogen Bronchogenic carcinoma, CML/CLL, lymphomas (lymphosarcoma , mycosis fungoides, polycythemia DNA Mechlorethamine vera, palliation Unclear Inhibitor of GSR Overexpression of GST isoenzymes [3,13,14] on HL III and IV). Also for the palliative treatment of metastatic carcinoma resulting in effusion. SUPPLEMENTARY TABLE 1. SUMMARY OF FDA-APPROVED ANTICANCER CYTOTOXIC DRUGS AT MAY 2019 (CONTINUATION) Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References Multiple myeloma, DNA Melphalan Unclear Inhibitor of SLC22A3 Bcl-X expression [3,15,16] ovarian Nitrogen mustard derivative mustard Nitrogen Anaplastic astrocytoma, DNA, Substrate of Carmustine glioblastoma Inhibitor of GSR Overexpression of MGMT [3,17–20] RNA CYP1A2 multiforme, multiple myeloma Glioma, STMN4, inhibitor of DNA Lomustine medulloblastoma, Unclear Overexpression of MGMT [3,21] CYP2D6, CYP3A4 lymphoma Nitrosurea Synthetic drugs Synthetic Alkylating Agents Alkylating SLC2A2 ligand, antagonist of Beta-n- Pancreas and acetylglucosaminida DNA Streptozocin carcinoid Unclear Overexpression of MGMT [22] se, MGEA5, inducer syndrome of ABCB1, CYP1A1, CYP1A2, CYP2E1. Substrate of Overexpression of MGMT, Bcl-2 expression, DNA, Melanoma, Dacarbazine CYP1A1, Inhibitor of PGD AKT activation, NF-kB activation, VEGFR-1 [3,23] POLA2 lymphomas CYP1A2, CYP expression Triazenes Anaplastic Overexpression of MGMT, Bcl-2 expression, Temozolomide astrocytoma, DNA Unclear Inducer of CYP3A4 AKT activation, NF-kB activation, VEGFR-1 [23,24] (Oral dacarbazine) glioblastoma expression multiforme SUPPLEMENTARY TABLE 1. SUMMARY OF FDA-APPROVED ANTICANCER CYTOTOXIC DRUGS AT MAY 2019 (CONTINUATION) Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References Substrate of ABCC2, ABCG2, Lung, ovarian ATP7A, ATP7B, Upregulation of P-type ATPases, and squamous GSTM1, GSTT1, SOD1, inducer of overexpression of MRP2, enhanced activity of DNA Carboplatin [3,25,26] cell head and GSTP1, MT1A, MPO, XDH, NER and overexpression of BRCA2, mutation neck MT2A, NQO1, and downregulation of RAD52 (repair gene) SLC31A1, SLC31A2 Bladder, cervical, Substrate of esophageal, ABCB1, ABCC6, A2M, ATOX1, MPG gastric, germ cell ABCG2, ATP7A, and TF. Inhibitor of tumours ATP7B, GSTM1, BCHE, CYP2B6, (including GSTP1, GSTT1, CYP2C9, nat, Upregulation of P-type ATPases, adducts testicular), DNA Cisplatin MT1A, MT2A, PTGS2 and overexpression of MRP2, enhanced activity of [3,25,27] mesothelioma, NQO1, SLC22A2. Inducer of NER and overexpression of BRCA1 NSCLC, SLC22A2, ABCC2, ABCC3, osteosarcoma, SLC31A1, ABCC5, CYP4A11, based agents based - ovarian, SCLC, SLC31A2 and MPO and XDH. and squamous SOD1. cell head and Synthetic drugs Synthetic neck Platinum Alkylating Agents / DNA / DNA Agents Alkylating Substrate of ATP7A, ATP7B, ABCC2, ABCG2, CYP1A1, CYP1B1, CYP2E1, CRC, Alterations in the mitochondrial apoptotic GSTM1, GSTT1, DNA Oxaliplatin esophageal and Unclear pathway, enhanced autophagy, increased GSH [3,28–30] GSTP1, MPO, gastric levels, SRBC hypermethylation (in vitro) MT1A, MT2A, NQO1, SLC22A2, SLC22A3, SLC31A1 and SOD1. SUPPLEMENTARY TABLE 1. SUMMARY OF FDA-APPROVED ANTICANCER CYTOTOXIC DRUGS AT MAY 2019 (CONTINUATION) Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References Substrate of ABCB1, ABCC1, ABCC2, ABCC3, ABCC4, ABCC11, ABCG2, AOX1, CYP3A4, Acute leukemias, DHFR, FPGS, bladder, brain, FOLR1, GGH, breast, Inhibitor of ABCC1, MTHFR, SLC16A1, Overexpression of ABC transporters, gestational ABCC2, ABCC3, SLC19A1, SLC22A6, downregulation of RFC, overexpression of DHFR Methotrexate trophoblastic ABCC4, ABCC10, [3,31,32] SLC22A7, SLC22A8, DHFR, loss of function of FPGS, disease, head PGD, SLC22A8, SLC22A11, overexpression of TS and neck, lung, SLC46A1 SLC46A1, NHL, SLCO1A2, osteosarcoma SLCO1B1, SLCO1B3, SLCO1C1, SLCO3A1, SLCO4C1, TYMS Antifolates Overexpression of ABC transporters, DHFR, Malignant Substrate of ATIC, inducer of downregulation of RFC, overexpression of GART, Pemetrexed mesothelioma [33] SLC22A8 DCK, SLC29A1 DHFR, loss of function of FPGS, TYMS and NSCLC Antimetabolite overexpression of TS Synthetic drugs Synthetic DHFR, T-cell Substrate of FPGS, decreased RFC-1 expression and increased Pralatrexate Unclear [34] TYMS Lymphoma SLC19A1 MDR1 expression DNA, RNA, 5-Azacitidine Leukemias Substrate of cytidine Unclear Mutations in TET2, overexpression of BCL2 [3,35] DNMT1 deaminase (CDA) analogs Nucleoside and precursor precursor and Nucleoside SUPPLEMENTARY TABLE 1. SUMMARY OF FDA-APPROVED ANTICANCER CYTOTOXIC DRUGS AT MAY 2019 (CONTINUATION) Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References ABCC3, ABCC4, ABCC5, ABCG2, CYP1A2, Anal, breast, CYP2A6, CRC, gastric, CYP2C8, DPYD, Overexpression of ABC transporters, DNA, RNA, 5-Fluorouracil head and neck, MTHFR, PPAT, Inhibitor of CYP2C9 overexpression of TS, enhanced activity of [3,36] TYMS esophageal and SLC22A7, DPD pancreas SLC29A1, TYMP, TYMS, UMPS, UPP1, UPP2 DNA, MGMT, PNP, POLA1, Hairy cell POLE2, leukemia, Substrate of Decreased expression of hENT1, reduced POLE3, Cladribine chronic DCK, ABCG2, Unclear activity of dCK, overexpression of 5'- [3,37] POLE4, lymphocytic SLC28A3. nucleotidase (in vitro) POLE, RRM1, leukemia RRM2, RRM2B. Acute lymphoblastic DNA, POLA1, Substrate of Clofarabine leukemia and Unclear Reduced activity of dCK [3,38] RRM1 DCK, ABCG2 acute myeloid leukemia Antimetabolite Acute myeloid Synthetic drugs Synthetic leukemia, acute lymphoblastic leukemia, Nucleoside and precursor analogs precursor and Nucleoside chronic myeloid leukemia, Substrate of Downregulation of dCK, downregulation of DNA, POLB Cytarabine lymphomas, CDA, CYP3A4, Unclear [3,39] hENT1 transporter progressive DCTD, NT5E multifocal leucoencephalop athy and meningeal leukemia Myelodysplastic syndrome, sickle cell anaemia (orphan), acute Substrate of Mutations in TET2, higher activity of CDA DNA, DNMT1 Decitabine Unclear [40,41] myeloid DCK combined with decreased activity of dCK leukemia and chronic myeloid leukemia. SUPPLEMENTARY TABLE 1. SUMMARY OF FDA-APPROVED ANTICANCER CYTOTOXIC DRUGS AT MAY 2019 (CONTINUATION) Classification Targets Name Indication Detox pathway Known off-targets Resistance mechanisms References Metastatic GI Floxuridine TYMS adenocarcinoma TYMP Inhibitor of CYP2C9 Amplification of TS and/or deletion of OPRTase [42–44] (5-FU analog) and gastric Acute myeloid leukemia, chronic DNA, lymphocytic Substrate of POLA1, Fludarabine leukemia, NHL SLC28A3, Agonist of DCK Mutations in TP53, overexpression of MYC [3,45,46] RRM1 and SLC29A1 Waldenstrom macroglobulinae mia. Bladder, breast, Substrate of nasopharyngeal, CDA, DCK, Antimetabolite Synthetic drugs Synthetic DNA, non-small cell ABCB1, RRM1, lung, ovarian ABCC10, Low expression of hENT1, higher dCDA Gemcitabine Unclear [3,47] TYMS, and pancreas, SLC28A1, expression, enhanced activity of dCK CMPK1 lymphomas and SLC28A3, Nucleoside and precursor analogs precursor and Nucleoside inflammatory SLC29A1, bowel disease. SLC29A2 Acute Substrate of lymphoblastic AOX1, TPMT, leukemia, acute HPRT1, XDH, SLC22A8, promyelocytic PPAT, ABCC4, ABCC5, Overexpression of ABC transporters, activating Mercaptopurine leukemia, Unclear [3,48] IMPDH1, SLC28A2, mutations in NT5C2 lymphoblastic IMPDH2 SLC28A3, lymphoma
Recommended publications
  • The TP53 Apoptotic Network Is a Primary Mediator of Resistance to BCL2 Inhibition in AML Cells
    Published OnlineFirst May 2, 2019; DOI: 10.1158/2159-8290.CD-19-0125 RESEARCH ARTICLE The TP53 Apoptotic Network Is a Primary Mediator of Resistance to BCL2 Inhibition in AML Cells Tamilla Nechiporuk1,2, Stephen E. Kurtz1,2, Olga Nikolova2,3, Tingting Liu1,2, Courtney L. Jones4, Angelo D’Alessandro5, Rachel Culp-Hill5, Amanda d’Almeida1,2, Sunil K. Joshi1,2, Mara Rosenberg1,2, Cristina E. Tognon1,2,6, Alexey V. Danilov1,2, Brian J. Druker1,2,6, Bill H. Chang2,7, Shannon K. McWeeney2,8, and Jeffrey W. Tyner1,2,9 ABSTRACT To study mechanisms underlying resistance to the BCL2 inhibitor venetoclax in acute myeloid leukemia (AML), we used a genome-wide CRISPR/Cas9 screen to identify gene knockouts resulting in drug resistance. We validated TP53, BAX, and PMAIP1 as genes whose inactivation results in venetoclax resistance in AML cell lines. Resistance to venetoclax resulted from an inability to execute apoptosis driven by BAX loss, decreased expression of BCL2, and/or reliance on alternative BCL2 family members such as BCL2L1. The resistance was accompanied by changes in mitochondrial homeostasis and cellular metabolism. Evaluation of TP53 knockout cells for sensitivities to a panel of small-molecule inhibitors revealed a gain of sensitivity to TRK inhibitors. We relate these observations to patient drug responses and gene expression in the Beat AML dataset. Our results implicate TP53, the apoptotic network, and mitochondrial functionality as drivers of venetoclax response in AML and suggest strategies to overcome resistance. SIGNIFICANCE: AML is challenging to treat due to its heterogeneity, and single-agent therapies have universally failed, prompting a need for innovative drug combinations.
    [Show full text]
  • 4Β-Hydroxycholesterol As Biomarker for Variation in CYP3A Activity
    ȕ-Hydroxycholesterol as biomarker for variation in CYP3A activity Dissertation for the Degree of Philosophiae Doctor (Ph.D.) Kristine Hole 2018 Center for Psychopharmacology Diakonhjemmet Hospital Oslo Department of Pharmaceutical Biosciences School of Pharmacy Faculty of Mathematics and Natural Sciences University of Oslo © Kristine Hole, 2018 Series of dissertations submitted to the Faculty of Mathematics and Natural Sciences, University of Oslo No. ISSN 1501-7710 All rights reserved. No part of this publication may be reproduced or transmitted, in any form or by any means, without permission. Cover: Hanne Baadsgaard Utigard. Print production: Reprosentralen, University of Oslo. TABLE OF CONTENTS ACKNOWLEDGEMENTS ...................................................................................................... II LIST OF PUBLICATIONS ..................................................................................................... III ABBREVIATIONS..................................................................................................................IV ABSTRACT.............................................................................................................................. V 1 INTRODUCTION.............................................................................................................. 1 1.1 Variability in drug response ....................................................................................... 1 1.2 Drug metabolism .......................................................................................................
    [Show full text]
  • Duvelisib Eliminates CLL B Cells, Impairs CLL- Supporting Cells, and Overcomes Ibrutinib Resistance in Preclinical Models
    Duvelisib Eliminates CLL B Cells, Impairs CLL- Supporting Cells, and Overcomes Ibrutinib Resistance in Preclinical Models Shih-Shih Chen ( [email protected] ) Karches Center for Oncology Research, the Feinstein Institutes for Medical Research Jacqueline Barrientos Karches Center for Oncology Research, the Feinstein Institutes for Medical Research Gerardo Ferrer Josep Carreras Leukaemia Research Institute (IJC) https://orcid.org/0000-0002-4084-6815 Priyadarshini Ravichandran Karches Center for Oncology Research, the Feinstein Institutes for Medical Research Michael Ibrahim Karches Center for Oncology Research, the Feinstein Institutes for Medical Research Yasmine Kieso Karches Center for Oncology Research, the Feinstein Institutes for Medical Research Jeff Kutok Innity Pharmaceuticals Marisa Peluso Innity Pharmaceuticals, Inc Sujata Sharma Innity Pharmaceuticals, Inc David Weaver Institute of Health Sciences, Anhui University Jonathan Pachter Verastem Inc. Kanti Rai The Karches Center for Oncology Research. The Feinstein Institutes for Medical Research. Nicholas Chiorazzi Karches Center for Oncology Research, the Feinstein Institutes for Medical Research Article Keywords: chronic lymphocytic leukemia, cancer therapy, Bruton’s Tyrosine Kinase (BTKi), phosphoinositide 3-kinase (PI3Ki) Page 1/23 Posted Date: July 21st, 2021 DOI: https://doi.org/10.21203/rs.3.rs-701669/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 2/23 Abstract Inhibitors of Bruton’s Tyrosine Kinase (BTKi) and phosphoinositide 3-kinase (PI3Ki) have signicantly improved therapy of chronic lymphocytic leukemia (CLL). However, the emergence of resistance to BTKi has introduced an unmet therapeutic need. Here we demonstrate in vitro and in vivo the essential roles of PI3K-δ for CLL B-cell survival and migration and of PI3K-γ in T-cell migration and macrophage polarization; and more ecacious inhibition in CLL-cell burden by dual inhibition of PI3K-δ,γ.
    [Show full text]
  • PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects
    International Journal of Molecular Sciences Review PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects Rosalin Mishra , Hima Patel, Samar Alanazi , Mary Kate Kilroy and Joan T. Garrett * Department of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267-0514, USA; [email protected] (R.M.); [email protected] (H.P.); [email protected] (S.A.); [email protected] (M.K.K.) * Correspondence: [email protected]; Tel.: +1-513-558-0741; Fax: +1-513-558-4372 Abstract: The phospatidylinositol-3 kinase (PI3K) pathway is a crucial intracellular signaling pathway which is mutated or amplified in a wide variety of cancers including breast, gastric, ovarian, colorectal, prostate, glioblastoma and endometrial cancers. PI3K signaling plays an important role in cancer cell survival, angiogenesis and metastasis, making it a promising therapeutic target. There are several ongoing and completed clinical trials involving PI3K inhibitors (pan, isoform-specific and dual PI3K/mTOR) with the goal to find efficient PI3K inhibitors that could overcome resistance to current therapies. This review focuses on the current landscape of various PI3K inhibitors either as monotherapy or in combination therapies and the treatment outcomes involved in various phases of clinical trials in different cancer types. There is a discussion of the drug-related toxicities, challenges associated with these PI3K inhibitors and the adverse events leading to treatment failure. In addition, novel PI3K drugs that have potential to be translated in the clinic are highlighted. Keywords: cancer; PIK3CA; resistance; PI3K inhibitors Citation: Mishra, R.; Patel, H.; Alanazi, S.; Kilroy, M.K.; Garrett, J.T.
    [Show full text]
  • Minutes of the CHMP Meeting 14-17 September 2020
    13 January 2021 EMA/CHMP/625456/2020 Corr.1 Human Medicines Division Committee for medicinal products for human use (CHMP) Minutes for the meeting on 14-17 September 2020 Chair: Harald Enzmann – Vice-Chair: Bruno Sepodes Disclaimers Some of the information contained in these minutes is considered commercially confidential or sensitive and therefore not disclosed. With regard to intended therapeutic indications or procedure scopes listed against products, it must be noted that these may not reflect the full wording proposed by applicants and may also vary during the course of the review. Additional details on some of these procedures will be published in the CHMP meeting highlights once the procedures are finalised and start of referrals will also be available. Of note, these minutes are a working document primarily designed for CHMP members and the work the Committee undertakes. Note on access to documents Some documents mentioned in the minutes cannot be released at present following a request for access to documents within the framework of Regulation (EC) No 1049/2001 as they are subject to on- going procedures for which a final decision has not yet been adopted. They will become public when adopted or considered public according to the principles stated in the Agency policy on access to documents (EMA/127362/2006). 1 Addition of the list of participants Official address Domenico Scarlattilaan 6 ● 1083 HS Amsterdam ● The Netherlands Address for visits and deliveries Refer to www.ema.europa.eu/how-to-find-us Send us a question Go to www.ema.europa.eu/contact Telephone +31 (0)88 781 6000 An agency of the European Union © European Medicines Agency, 2020.
    [Show full text]
  • PRIOR AUTHORIZATION CRITERIA for APPROVAL Initial Evaluation Target Agent(S) Will Be Approved When ONE of the Following Is Met: 1
    Self-Administered Oncology Agents Through Preferred Prior Authorization Program Summary FDA APPROVED INDICATIONS3-104 Please reference individual agent product labeling. CLINICAL RATIONALE For the purposes of the Self -Administered Oncology Agents criteria, indications deemed appropriate are those approved in FDA labeling and/or supported by NCCN Drugs & Biologics compendia with a category 1 or 2A recommendation, AHFS, or DrugDex with level of evidence of 1 or 2A. SAFETY3-104 Agent(s) Contraindication(s) Afinitor/Afinitor Disperz Hypersensitivity to everolimus, to other rapamycin (everolimus) derivatives None Alecensa (alectinib) Alunbrig (brigatinib) None Ayvakit (avapritinib) None Balversa (erdafitinib) None Hypersensitivity to bosutinib Bosulif (bosutinib) Braftovi (encorafenib) None Brukinsa (zanubrutinib) None Cabometyx None (cabozantinib) Calquence None (acalabrutinib) Caprelsa Congenital long QT syndrome (vandetanib) Cometriq None (cabozantinib) Copiktra (duvelisib) None Cotellic (cobimetinib) None Daurismo (glasdegib) None None Erivedge (vismodegib) Erleada (apalutamide) Pregnancy None Farydak (panobinostat) Fotivda (tivozanib) None Gavreto (pralsetinib) None None Gilotrif (afatinib) Gleevec None (imatinib) Hycamtin Severe hypersensitivity to topotecan (topotecan) None Ibrance (palbociclib) KS_PS_SA_Oncology_PA_ProgSum_AR1020_r0821v2 Page 1 of 19 © Copyright Prime Therapeutics LLC. 08/2021 All Rights Reserved Effective: 10/01/2021 Agent(s) Contraindication(s) None Iclusig (ponatinib) Idhifa (enasidenib) None Imbruvica (ibrutinib)
    [Show full text]
  • Cytochrome P450 Enzymes in Oxygenation of Prostaglandin Endoperoxides and Arachidonic Acid
    Comprehensive Summaries of Uppsala Dissertations from the Faculty of Pharmacy 231 _____________________________ _____________________________ Cytochrome P450 Enzymes in Oxygenation of Prostaglandin Endoperoxides and Arachidonic Acid Cloning, Expression and Catalytic Properties of CYP4F8 and CYP4F21 BY JOHAN BYLUND ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2000 Dissertation for the Degree of Doctor of Philosophy (Faculty of Pharmacy) in Pharmaceutical Pharmacology presented at Uppsala University in 2000 ABSTRACT Bylund, J. 2000. Cytochrome P450 Enzymes in Oxygenation of Prostaglandin Endoperoxides and Arachidonic Acid: Cloning, Expression and Catalytic Properties of CYP4F8 and CYP4F21. Acta Universitatis Upsaliensis. Comprehensive Summaries of Uppsala Dissertations from Faculty of Pharmacy 231 50 pp. Uppsala. ISBN 91-554-4784-8. Cytochrome P450 (P450 or CYP) is an enzyme system involved in the oxygenation of a wide range of endogenous compounds as well as foreign chemicals and drugs. This thesis describes investigations of P450-catalyzed oxygenation of prostaglandins, linoleic and arachidonic acids. The formation of bisallylic hydroxy metabolites of linoleic and arachidonic acids was studied with human recombinant P450s and with human liver microsomes. Several P450 enzymes catalyzed the formation of bisallylic hydroxy metabolites. Inhibition studies and stereochemical analysis of metabolites suggest that the enzyme CYP1A2 may contribute to the biosynthesis of bisallylic hydroxy fatty acid metabolites in adult human liver microsomes. 19R-Hydroxy-PGE and 20-hydroxy-PGE are major components of human and ovine semen, respectively. They are formed in the seminal vesicles, but the mechanism of their biosynthesis is unknown. Reverse transcription-polymerase chain reaction using degenerate primers for mammalian CYP4 family genes, revealed expression of two novel P450 genes in human and ovine seminal vesicles.
    [Show full text]
  • RESEARCH ARTICLE Lobaplatin Combined Floxuridine/Pirarubicin
    DOI:http://dx.doi.org/10.7314/APJCP.2014.15.5.2057 The Effect Observation of LBP-based TACE for Unresectable Primary Hepatocellular Carcinoma RESEARCH ARTICLE Lobaplatin Combined Floxuridine/Pirarubicin-based Transcatheter Hepatic Arterial Chemoembolization for Unresectable Primary Hepatocellular Carcinoma Chang Zhao1, Xu-Jie Wang2*, Song Wang2*, Wei-Hua Feng2, Lei Shi2, Chun- Peng Yu2 Abstract Purpose: To assess the effect and safety of lobaplatin combinated floxuridine /pirarubicin in transcatheter hepatic arterial chemoembolization(TACE) of unresectable primary liver cancer. Patients and Methods: TACE combined with the chemotherapy regimen was used to treat 34 unresectable primary liver cancer patients. DSA/ MRI/CT/blood routine examinations were used to evaluate short term activity and toxicity after 4-5 weeks, the process being repeated if necessary. Results: Among the 34 cases, 1 (2.9%) showed a complete response, 21 (61.7%) a partial response, 8 (23.5%) stable disease, and 4 progressive disease, with a total effective rate of 67.6%. The content of alpha fetoprotein dropped by over 50% in 20 cases (58.8%). The rate of recovery was hepatalgia (88.2%), ascites (47.1%), appetite (55.9%), Performance Status(30.4%). The median follow-up time (MFT) was 281 days (63-558 days), and median progression-free survival was 118.5 days (95%, CI:88.8-148.2days). Adverse reactions (III-IV grade) were not common, with only 4 cases of vomiting and 2 cases of thrombocytopenia (III grade). Conclusions: Lobaplatin-based TACE is an effective and safe treatment for primary liver cancer. Keywords: Primary hepatic carcinoma - chemoembolization - platinum chemotherapy - clinical response Asian Pac J Cancer Prev, 15 (5), 2057-2060 Introduction LBP-based TACE, male 29 cases, female 5 cases, average age for 57.2±10.7 years, 12 cases were biopsy-proven, Hepatocellular carcinoma (HCC) is a common 22 cases were in line with the China Cancer Association malignant tumor .
    [Show full text]
  • Pharmacogenomic Characterization in Bipolar Spectrum Disorders
    pharmaceutics Review Pharmacogenomic Characterization in Bipolar Spectrum Disorders Stefano Fortinguerra 1,2 , Vincenzo Sorrenti 1,2,3 , Pietro Giusti 2, Morena Zusso 2 and Alessandro Buriani 1,2,* 1 Maria Paola Belloni Center for Personalized Medicine, Data Medica Group (Synlab Limited), 35131 Padova, Italy; [email protected] (S.F.); [email protected] (V.S.) 2 Department of Pharmaceutical & Pharmacological Sciences, University of Padova, 35131 Padova, Italy; [email protected] (P.G.); [email protected] (M.Z.) 3 Bendessere™ Study Center, Solgar Italia Multinutrient S.p.A., 35131 Padova, Italy * Correspondence: [email protected] Received: 25 November 2019; Accepted: 19 December 2019; Published: 21 December 2019 Abstract: The holistic approach of personalized medicine, merging clinical and molecular characteristics to tailor the diagnostic and therapeutic path to each individual, is steadily spreading in clinical practice. Psychiatric disorders represent one of the most difficult diagnostic challenges, given their frequent mixed nature and intrinsic variability, as in bipolar disorders and depression. Patients misdiagnosed as depressed are often initially prescribed serotonergic antidepressants, a treatment that can exacerbate a previously unrecognized bipolar condition. Thanks to the use of the patient’s genomic profile, it is possible to recognize such risk and at the same time characterize specific genetic assets specifically associated with bipolar spectrum disorder, as well as with the individual response to the various therapeutic options. This provides the basis for molecular diagnosis and the definition of pharmacogenomic profiles, thus guiding therapeutic choices and allowing a safer and more effective use of psychotropic drugs. Here, we report the pharmacogenomics state of the art in bipolar disorders and suggest an algorithm for therapeutic regimen choice.
    [Show full text]
  • Generated by SRI International Pathway Tools Version 25.0, Authors S
    An online version of this diagram is available at BioCyc.org. Biosynthetic pathways are positioned in the left of the cytoplasm, degradative pathways on the right, and reactions not assigned to any pathway are in the far right of the cytoplasm. Transporters and membrane proteins are shown on the membrane. Periplasmic (where appropriate) and extracellular reactions and proteins may also be shown. Pathways are colored according to their cellular function. Gcf_000238675-HmpCyc: Bacillus smithii 7_3_47FAA Cellular Overview Connections between pathways are omitted for legibility.
    [Show full text]
  • Synonymous Single Nucleotide Polymorphisms in Human Cytochrome
    DMD Fast Forward. Published on February 9, 2009 as doi:10.1124/dmd.108.026047 DMD #26047 TITLE PAGE: A BIOINFORMATICS APPROACH FOR THE PHENOTYPE PREDICTION OF NON- SYNONYMOUS SINGLE NUCLEOTIDE POLYMORPHISMS IN HUMAN CYTOCHROME P450S LIN-LIN WANG, YONG LI, SHU-FENG ZHOU Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P. R. China (LL Wang & Y Li) Discipline of Chinese Medicine, School of Health Sciences, RMIT University, Bundoora, Victoria 3083, Australia (LL Wang & SF Zhou). 1 Copyright 2009 by the American Society for Pharmacology and Experimental Therapeutics. DMD #26047 RUNNING TITLE PAGE: a) Running title: Prediction of phenotype of human CYPs. b) Author for correspondence: A/Prof. Shu-Feng Zhou, MD, PhD Discipline of Chinese Medicine, School of Health Sciences, RMIT University, WHO Collaborating Center for Traditional Medicine, Bundoora, Victoria 3083, Australia. Tel: + 61 3 9925 7794; fax: +61 3 9925 7178. Email: [email protected] c) Number of text pages: 21 Number of tables: 10 Number of figures: 2 Number of references: 40 Number of words in Abstract: 249 Number of words in Introduction: 749 Number of words in Discussion: 1459 d) Non-standard abbreviations: CYP, cytochrome P450; nsSNP, non-synonymous single nucleotide polymorphism. 2 DMD #26047 ABSTRACT Non-synonymous single nucleotide polymorphisms (nsSNPs) in coding regions that can lead to amino acid changes may cause alteration of protein function and account for susceptivity to disease. Identification of deleterious nsSNPs from tolerant nsSNPs is important for characterizing the genetic basis of human disease, assessing individual susceptibility to disease, understanding the pathogenesis of disease, identifying molecular targets for drug treatment and conducting individualized pharmacotherapy.
    [Show full text]
  • Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with BRCA1/2- and Non–BRCA1/2-Mutant Cancers
    Published OnlineFirst June 12, 2020; DOI: 10.1158/2159-8290.CD-20-0163 RESEARCH ARTICLE Phase I Trial of the PARP Inhibitor Olaparib and AKT Inhibitor Capivasertib in Patients with BRCA1/2- and Non–BRCA1/2-Mutant Cancers Timothy A. Yap1,2, Rebecca Kristeleit3, Vasiliki Michalarea1, Stephen J. Pettitt4,5, Joline S.J. Lim1, Suzanne Carreira2, Desamparados Roda1,2, Rowan Miller3, Ruth Riisnaes2, Susana Miranda2, Ines Figueiredo2, Daniel Nava Rodrigues2, Sarah Ward1,2, Ruth Matthews1,2, Mona Parmar1,2, Alison Turner1,2, Nina Tunariu1, Neha Chopra1,4, Heidrun Gevensleben2, Nicholas C. Turner1,4, Ruth Ruddle2, Florence I. Raynaud2, Shaun Decordova2, Karen E. Swales2, Laura Finneran2, Emma Hall2, Paul Rugman6, Justin P.O. Lindemann6, Andrew Foxley6, Christopher J. Lord4,5, Udai Banerji1,2, Ruth Plummer7, Bristi Basu8, Juanita S. Lopez1,2, Yvette Drew7, and Johann S. de Bono1,2 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst June 12, 2020; DOI: 10.1158/2159-8290.CD-20-0163 ABSTRACT Preclinical studies have demonstrated synergy between PARP and PI3K/AKT path- way inhibitors in BRCA1 and BRCA2 (BRCA1/2)–deficient andBRCA1/2 -proficient tumors. We conducted an investigator-initiated phase I trial utilizing a prospective intrapatient dose- escalation design to assess two schedules of capivasertib (AKT inhibitor) with olaparib (PARP inhibi- tor) in 64 patients with advanced solid tumors. Dose expansions enrolled germline BRCA1/2-mutant tumors, or BRCA1/2 wild-type cancers harboring somatic DNA damage response (DDR) or PI3K–AKT pathway alterations. The combination was well tolerated. Recommended phase II doses for the two schedules were: olaparib 300 mg twice a day with either capivasertib 400 mg twice a day 4 days on, 3 days off, or capivasertib 640 mg twice a day 2 days on, 5 days off.
    [Show full text]