Antineoplastic agents
INN Reviews Chemical classification Preg. cat. Route[1] Mechanism of action[1][2][3][4] Indications[1][2][4] Major toxicities[1][2][4][5]
1. Cytotoxic antineoplastics
1.01 Nucleoside analogues
Azacitidine [6][7][8][9]
[10][11][12]
[13][14][15]
Cytidine analogue X (Au) SC, IV DNA methyltransferase inhibitor and incorporates itself into RNA, hence inhibiting gene expression.[16] Myelodysplastic syndromes, acute myeloid leukaemiaand chronic myeloid leukaemia Myelosuppression, kidney failure (uncommon/rare), renal tubular acidosis andhypokalaemia.
Capecitabine [17][18][19]
[20][21][22]
[23][24][25]
Uracil analogue D (Au) PO Fluorouracil prodrug Breast, colorectal, gastric and oesophageal cancer Myelosuppression, cardiotoxicity, hypertriglyceridaemia, GI haemorrhage (uncommon), cerebellar syndrome (uncommon), encephalopathy (uncommon) and diarrhoea.
Carmofur N/A Uracil analogue N/A PO Fluorouracil prodrug Colorectal, breast and ovarian cancer Myelosuppression, neurotoxicity and diarrhoea.
Cladribine MS:
[26][27][28]
[29][30][31]
[32][33][34]
[35]
Cancer:
[36][37][38]
[39]
Adenosine analogue D (Au) PO, SC, IV DNA methyltransferaseinhibitor, metabolites incorporate themselves into DNA.[40] Hairy cell leukaemia, chronic lymphocytic leukaemia, Waldenstrom macroglobulinaemia and multiple sclerosis. Myelosuppression,haemolytic anaemia (uncommon), neurotoxicity (rare), renal impairment (rare), pulmonary interstitial infiltrates (rare), Stevens–Johnson syndrome (rare) and toxic epidermal necrolysis (rare).
Clofarabine [41][42][43]
[44][45][46]
[47][48][49]
Deoxyadenosine analogue D (Au) IV Ribonucleotide reductase and DNA polymerase inhibitor.[50] Acute lymphoblastic leukaemia andacute myeloid leukaemia Myelosuppression, hypokalaemia, cytokine release syndrome, Stevens–Johnson syndrome(uncommon), toxic epidermal necrolysis (uncommon) and pancreatitis (uncommon)
Cytarabine [51][52][53] Cytidine analogue D (Au) SC, IM, IV, IT DNA polymerase inhibitor, S-phase specific. Incorporates its metabolites into DNA. Acute myeloid leukaemia, acute lymphoblastic leukaemia, chronic myeloid leukaemia, lymphomas,progressive multifocal leucoencephalopathy and meningeal leukaemia Myelosuppression, GI bleeds, pancreatitis (uncommon/rare),anaphylaxis (uncommon/rare), pericarditis (uncommon/rare) and conjunctivitis (uncommon/rare). High dose: cerebral and cerebellar dysfunction, ocular toxicity, pulmonary toxicity, severe GI ulceration and peripheral neuropathy (rare).
Decitabine [6][7][54]
[55][56][57]
[58][59][60]
[61][62]
Cytidine analogue. D (US) IV DNA methyltransferase inhibitor. Myelodysplastic syndrome, sickle cell anaemia (orphan), acute myeloid leukaemia and chronic myeloid leukaemia. Myelosuppression, hyperglycaemia, hypoalbuminaemia, hypomagnesaemia, hypokalaemia, hyperkalaemia and thrombocythaemia.
Floxuridine [63] Uracil analogue D (US) IA Fluorouracil analogue. Metastatic GI adenocarcinoma and stomach cancer Myelosuppression.
Fludarabine [64][65][66]
[67][68][69]
[70]
Adenosine analogue D (Au) PO, IV DNA polymerase and ribonucleotide reductase inhibitor. Acute myeloid leukaemia, chronic lymphocytic leukaemia, non-Hodgkin lymphoma and Waldenstrom macroglobulinaemia. Myelosuppression,hyperglycaemia, GI bleeds (uncommon), pneumonitis (uncommon), haemolytic anaemia (uncommon), severe neurotoxicity (rare),haemorrhagic cystitis (rare), Stevens–Johnson syndrome (rare) and toxic epidermal necrolysis (rare).
Fluorouracil [71][72][73] Uracil analogue D (Au) IV Thymidylate synthase inhibitor. Anal, breast, colorectal, gastric, head and neck, oesophageal and pancreatic cancer. Bowen's disease and actinic keratoses. Myelosuppression, diarrhoea, cardiotoxicity, GI ulceration and bleeding (uncommon), cerebellar syndrome (uncommon), encephalopathy (uncommon) and anaphylaxis (rare).
Gemcitabine [74][75][76]
[77][78][79]
[80]
Deoxycytidine analogue D (Au) IV DNA synthesis inhibitor, induces apoptosis specifically in S-phase. Bladder, breast, nasopharyngeal,non-small cell lung, ovarian and pancreatic cancer, lymphomas andinflammatory bowel disease. Myelosuppression, pulmonary toxicity, kidney failure (rare), haemolytic uraemic syndrome(rare), thrombotic thrombocytopenic purpura (rare), anaphylactoid reaction (rare), reversible posterior leucoencephalopathy syndrome (rare), myocardial infarction (rare) and heart failure (rare).
Mercaptopurine [81][82] Hypoxanthine analogue D (Au) PO Purine synthesis inhibitor. Acute lymphoblastic leukaemia, acute promyelocytic leukaemia, lymphoblastic lymphoma and inflammatory bowel disease.[83] Myelosuppression, hepatotoxicity, GI ulceration (rare), pancreatitis (rare) and secondary leukaemia (rare) or myelodysplasia (rare).
Nelarabine[84] [85][86]
[87][88][89]
[90][91]
Adenosine analogue D (US) IV Purine synthesis inhibitor. Acute lymphoblastic leukaemia and chronic lymphocytic leukaemia. Myelosuppression, pleural effusion, seizures, tumour lysis syndrome and a condition similar to Guillain-Barré syndrome.
Pentostatin [92][93][94]
[95][96][97]
Adenosine analogue D (US) IV Adenosine deaminase inhibitor. Hairy cell leukaemia, peripheral T-cell lymphoma (orphan), cutaneous T cell lymphoma (orphan) and chronic lymphocytic leukaemia (orphan). Myelosuppression, neurotoxicity, immune hypersensitivity, hyponatraemia, thrombotic thrombocytopenic purpura and microangiopathic hemolytic anaemia.
Tegafur [98][99][100]
[101][102][103]
[104]
Fluorouracilanalogue D (Au) PO Thymidylate synthase inhibitor. Breast, colorectal cancer, gallbladder, gastrointestinal tract, head and neck, liver and pancreas cancer. Myelosuppression, diarrhoea, neurotoxicity and hepatitis (rare).
Tioguanine [105][106][107]
[108][109]
Guanine analogue. D (Au) PO Purine synthesis inhibitor. Acute lymphoblastic leukaemia and acute myeloid leukaemia Myelosuppression, hepatotoxicity, peripheral neuropathy (uncommon), intestinal necrosis (rare) and perforation (rare).
1.02 Antifolates
Methotrexate [110][111][112]
[113][114][115]
Folate analogue D (Au) SC, IM, IV, IT, PO Dihydrofolate reductase inhibitor. Bladder and breast cancer. squamous cell carcinoma of head and neck, gestational trophoblastic disease, acute leukaemias, non-Hodgkin lymphoma, osteosarcoma, brain tumours, graft-versus-host disease and systemic sclerosis. Myelosuppression, pulmonary toxicity, hepatotoxicity, neurotoxicity (high dose or intrathecal administration), anaphylactic reactions (rare), Stevens–Johnson syndrome (rare), Toxic Epidermal Necrolysis (rare), kidney failure (rare), osteoporosis (rare), skin and bone necrosis (rare) and macrocytic anaemia (rare).
Pemetrexed [116][117][118]
[119][120][121]
[122][123]
[124][125][126]
[127][128][129]
[130][131][132]
[133][134][135]
[136][137][138]
[139][140][141]
Folate analogue D (Au) IV Dihydrofolate reductase, thymidylate synthase and glycinamide ribonucleotide formyltransferase inhibitors. Malignant mesothelioma and non-squamous non-small cell lung cancer. Myelosuppression, renal impairment, peripheral neuropathy, Supraventricular tachycardia (uncommon), hepatitis (rare), colitis (rare), pneumonitis (rare), radiation recall (rare), Stevens–Johnson syndrome (rare) and toxic epidermal necrolysis (rare).
Raltitrexed [142][143][144]
[145][146][147]
[148][149]
Quinazolinone D (Au) IV Dihydrofolate reductase and thymidylate synthase inhibitor. Colorectal cancer Myelosuppression
1.03 Other antimetabolites
Hydroxycarbamide [150][151][152]
[153][154][155]
[156][157][158]
[159]
Urea analogue D (Au) PO Inhibits DNA synthesis by inhibiting the enzyme ribonucleotide reductase. Chronic myeloid leukaemia, essential thrombocytosis, polycythaemia vera, myelofibrosis, acute myeloid leukaemia and sickle cell anaemia Myelosuppression, skin cancer (rare), oedema (rare), hallucinations (rare), seizures (rare) and pulmonary toxicity (rare).
1.04 Topoisomerase I inhibitor
Irinotecan Camptothecin D (Au) IV Inhibits topoisomerase I. Colorectal cancer Diarrhoea, myelosuppression, pulmonary infiltrates (uncommon), bradycardia (uncommon), ileus (rare) and colitis (rare).
Topotecan Camptothecin D (Au) IV Inhibits topoisomerase I. Small cell lung cancer, ovarian cancer and cervical cancer Diarrhoea, myelosuppression, interstitial lung disease and allergy.
1.05 Anthracyclines
Daunorubicin Anthracycline D (Au) IV Inhibits DNA and RNA synthesis by intercalating DNA base pairs. Inhibits DNA repair by inhibiting topoisomerase II. Acute leukaemias Myelosuppression, cardiotoxicity, anaphylaxis (rare), secondary malignancies (particularly acute myeloid leukaemia and myelodysplastic syndrome) and radiation recall.
Doxorubicin Anthracycline D (Au) IV As above. Breast cancer, lymphomas, sarcomas, bladder cancer, acute lymphoblastic leukaemia, Wilms' tumour, AIDS-related Kaposi's sarcoma, neuroblastoma and multiple myeloma As above.
Epirubicin Anthracycline D (Au) IV As above. Breast cancer, gastric cancer and bladder cancer As above.
Idarubicin Anthracycline D (Au) IV, PO As above. Acute leukaemias. As above.
Mitoxantrone Anthracenedione D (Au) IV As above. Non-Hodgkin lymphoma, acute myeloid leukaemia,prostate cancer and multiple sclerosis As above.
Valrubicin Anthracycline C (US) IV As above. Bladder cancer. As above.
1.06 Podophyllotoxins
Etoposide Podophyllotoxin D (Au) IV, PO Topoisomerase II inhibitor. Testicular cancer, ovarian cancer, lung cancer, acute myeloid leukaemia, lymphomas and sarcomas Myelosuppression, hypersensitivity reactions, Stevens–Johnson syndrome (rare), peripheral neuropathy (uncommon) and secondary malignancies (especially acute myeloid leukaemia).
Teniposide Podophyllotoxin D (Au) IV Topoisomerase II inhibitor. Lymphomas, acute lymphoblastic leukaemia and neuroblastoma As above.
1.07 Taxanes
Cabazitaxel Taxane D (Au) IV Microtubule disassembly inhibitor. Arrests cells in late G2 phase and M phase. Prostate cancer Myelosuppression, diarrhoea, kidney failure, hypersensitivity, severe GI reactions (including perforation, ileus, colitis, etc.; all rare) and peripheral neuropathy
Docetaxel Taxane D (Au) IV As above. Breast cancer, non-small cell lung cancer, ovarian cancer, prostate cancer, squamous cell head and neck cancer and gastric cancer. Myelosuppression, peripheral neuropathy, hypersensitivity, fluid retention, heart failure (uncommon), pulmonary toxicity (rare), radiation recall (rare), scleroderma-like skin changes (rare), Stevens–Johnson syndrome (rare), toxic epidermal necrolysis (rare), seizures (rare) and encephalopathy (rare)
Paclitaxel Taxane D (Au) IV As above. Ovarian cancer, breast cancer, non-small cell lung cancer, AIDS-related Kaposi's sarcoma, cervical cancer, germ cell cancer and endometrial cancer Hypersensitivity, myelosuppression, peripheral neuropathy, myocardial infarction (uncommon), arrhythmias (uncommon), pulmonary toxicity (rare), radiation recall (rare), scleroderma-like skin changes (rare), Stevens–Johnson syndrome (rare), toxic epidermal necrolysis (rare), seizures (rare) and encephalopathy (rare).
1.08 Vinca alkaloids
Vinblastine Vinca alkaloid D (Au) IV Microtubule assembly inhibitor. Arrests cells in M phase. Hodgkin lymphoma, germ cell tumours, non-small cell lung cancer, bladder cancer and primary immune thrombocytopenia Neurotoxicity, myelosuppression, myocardial ischaemia (rare) and myocardial infarction (rare).
Vincristine Vinca alkaloid D (Au) IV As above. Lymphomas, acute lymphoblastic leukaemia, multiple myeloma, sarcoma, brain tumours, Wilms' tumour, neuroblastoma and primary immune thrombocytopenia Neurotoxicity, anaphylaxis (rare), myocardial ischaemia (rare) and myocardial infarction (rare).
Vindesine Vinka alkaloid D (Au) IV As above. Refractory metastatic melanoma, childhood acute lymphoblastic leukaemia, chronic myeloid leukaemia in blast crises, neuroblastoma, non-small cell lung cancer and breast cancer. Myelosuppression, neurotoxicity and paralytic ileus.
Vinflunine Vinca alkaloid D (Au) IV As above. Bladder cancer As per vinblastine.
Vinorelbine Vinca alkaloid D (Au) IV As above. Breast cancer and non-small cell lung cancer. As above.
1.09 Alkylating agents
Bendamustine Nitrogen mustard D (Au) IV Alkylates DNA. Chronic lymphocytic leukaemia, mantle cell lymphoma and non-Hodgkin's lymphoma. Myelosuppression, hypokalaemia and tachycardia.
Busulfan Dialkylsulfonate D (Au) IV, PO Alkylates DNA. Conditioning treatment before haematopoietic stem cell transplantation (high dose, IV), chronic myeloid leukaemia, myelofibrosis, polycythaemia vera and essential thrombocytosis Myelosuppression, seizures (high dose), tachycardia (high dose), hepatic sinusoidal obstruction syndrome (high dose), Addison-like syndrome (rare), pulmonary fibrosis (rare), cataracts (rare) and hepatitis (rare). Secondary malignancies.[1][160]
Carmustine Nitrosourea D (Au) IV Alkylates DNA. Anaplastic astrocytoma, glioblastoma multiforme andmycosis fungoides (topical) Myelosuppression, pulmonary fibrosis, pulmonary infiltrates, seizure, brain oedema, cerebrospinal leaks, subdural fluid collection, intracranial infection, hypotension (uncommon), tachycardia (uncommon), decrease in kidney size (reversible), uraemia (uncommon), kidney failure (uncommon), severe hepatic toxicity (rare), thrombosis (rare) and neuroretinitis (rare). Secondary malignancies.[1][160]
Chlorambucil Nitrogen mustard D (Au) IV Alkylates DNA. Lymphoma, chronic lymphocytic leukaemia andWaldenström's macroglobulinaemia Myelosuppression, hallucinations (rare), seizures (rare), sterile cystitis (rare), hepatotoxicity (rare), severe pneumonitis (rare), Stevens–Johnson syndrome (rare), toxic epidermal necrolysis (rare) and drug fever (rare). Secondary malignancies.[1][160]
Chlormethine Nitrogen mustard D (Au) IV, topical Alkylates DNA. Cutaneous T-Cell Lymphoma, metastatic carcinoma, leukaemias, lymphomas, polycythemia vera and bronchogenic carcinoma Thrombosis, myelosuppression (common), hyperuricaemia, erythema multiforme, haemolytic anaemia, nausea and vomiting (severe) and secondary malignancies.[160]
Cyclophosphamide Nitrogen mustard D (Au) IV Alkylates DNA. Breast cancer, lymphoma, acute lymphoblastic leukaemia, chronic lymphocytic leukaemia, sarcoma, multiple myeloma, Waldenström's macroglobulinaemia, systemic lupus erythematosus, glomerulonephritis, systemic vasculitis and Wegener's granulomatosis Myelosuppression, nausea and vomiting (>30%), haemorrhagic cystitis, heart failure (rare), pulmonary fibrosis (rare), hepatic sinusoidal obstruction syndrome (rare), water retention resembling SIADH (rare) and seizures (rare). Secondary malignancies.[160]
Dacarbazine Triazene D (Au) IV Alkylates DNA. Hodgkin lymphoma, metastatic malignant melanoma and soft tissue sarcoma Myelosuppression, agranulocytosis (uncommon), hepatic vein thrombosis (rare) and hepatocellular necrosis (rare). Secondary malignancies.[160]
Fotemustine Nitrosourea D (Au) IV Alkylates DNA. Metastatic malignant melanoma. Myelosuppression.
Ifosfamide Nitrogen mustard D (Au) IV Alkylates DNA. Sarcomas, testicular cancer and lymphomas. Myelosuppression, haemorrhagic cystitis, nephrotoxicity, neurotoxicity and cardiac toxicity (rare). Secondary malignancies.[160]
Lomustine Nitrosourea D (Au) PO Alkylates DNA. Glioma and medulloblastoma. Myelosuppression, pulmonary infiltration and fibrosis. Secondary malignancies.[160]
Melphalan Nitrogen mustard D (Au) IV, PO Alkylates DNA. Malignant melanoma of the extremities, multiple myeloma, conditioning treatment before haemopoietic stem cell transplant. Myelosuppression, pulmonary fibrosis and pneumonitis (uncommon), skin necrosis (uncommon), anaphylaxis, hepatic sinusoidal obstruction syndrome and SIADH. Secondary malignancies.[160]
Streptozotocin Nitrosourea D (Au) IV, PO Alkylates DNA. Pancreatic cancer and carcinoid syndrome. Nephrotoxicity, hypoglycaemia, myelosuppression, nausea and vomiting (>90%), jaundice and nephrogenic diabetes insipidus (rare).
Temozolomide Triazene D (US) PO Alkylates DNA. Anaplastic astrocytoma, glioblastoma multiforme, metastatic malignant melanoma Myelosuppression, Stevens–Johnson syndrome (rare), pneumonitis (rare) and hepatitis (rare).
1.10 Platinum compounds
Carboplatin Platinum complex D (Au) IV Reacts with DNA, inducing apoptosis, non-cell cycle specific. Ovarian cancer, lung cancer and squamous cell head and neck cancer Myelosuppression, nausea and vomiting (30-90%), peripheral neuropathy, ototoxicity, anaphylaxis, acute kidney failure (rare), haemolytic uraemic syndrome (rare) and loss of vision (rare).
Cisplatin Platinum complex D (Au) IV As above. Germ cell tumours (including testicular cancer), ovarian cancer, cervical cancer, small cell and non-small cell lung cancer, mesothelioma, squamous cell head and neck cancer, oesophageal cancer, gastric cancer, bladder cancer and osteosarcoma Nephrotoxicity, nausea and vomiting (30-100%), myelosuppression, electrolyte anomalies, peripheral neuropathy, ototoxicity and anaphylaxis, haemolytic anaemia (rare), optic neuritis (rare), reversible posterior leucoencephalopathy syndrome (rare), seizures (rare), ECG changes (rare) and heart failure (rare).
Nedaplatin [161] Platinum complex N/A IV As above. Non-small cell lung cancer, oesophageal cancer, uterine cervical cancer, head and neck cancer and urothelial cancer Nephrotoxicity, myelosuppression and nausea and vomiting (30-90%).
Oxaliplatin Platinum complex D (Au) IV As above. Colorectal cancer, oesophageal cancer and gastric cancer Myelosuppression, peripheral neuropathy, anaphylaxis, nausea and vomiting (30-90%), hypokalaemia, metabolic acidosis, interstitial lung disease (uncommon), ototoxicity (rare), reversible posterior leucoencephalopathy syndrome (rare), immune-mediated cytopenias (rare) and hepatic sinusoidal obstruction syndrome (rare).
1.11 Miscellaneous others
Altretamine Triazine D (Au) PO Unclear, reactive intermediates covalently bind to microsomal proteins and DNA, possibly causing DNA damage Recurrent ovarian cancer Myelosuppression, peripheral neuropathy, seizures and hepatotoxicity (rare).
Bleomycin Glycopeptide D (Au) IM, SC, IA, IV or IP Inhibits DNA and to a lesser extent RNA synthesis, produces single and double strand breaks in DNA possibly by free radical formation. Germ cell tumours, squamous cell carcinoma, pancreatic cancer, non-Hodgkin's, pleural sclerosing and Hodgkin's lymphoma. Pulmonary toxicity, hypersensitivity, scleroderma andRaynaud's phenomenon.
Bortezomib Dipeptidyl boronic acid C (Au) IV, SC Proteasome inhibitor. Multiple myeloma, mantle cell lymphoma and follicular lymphoma (orphan). Peripheral neuropathy, neutropenia, thrombocytopenia, anaemia, orthostatic hypotension, hepatitis (uncommon/rare), haemorrhage (uncommon/rare), heart failure (uncommon/rare), seizures (uncommon/rare), progressive multifocal leucoencephalopathy (PML) and hearing loss.
Dactinomycin Polypeptide D (Au) IV Complexes with DNA interfering with DNA-dependent RNA synthesis Gestational trophoblastic disease, Wilms' tumour and rhabdomyosarcoma Myelosuppression, anaphylaxis, radiation recall, hepatotoxicity and hepatic sinusoidal obstruction syndrome (common in Wilms' tumour).
Estramustine Nitrogen mustard and oestrogen analogue D (Au) PO Antimicrotubule and oestrogenic actions Prostate cancer. Cardiovascular complications, such as ischaemic heart disease, venous thromboembolism, congestive heart failure, pulmonary embolism, myocardial infarction and cerebrovascular failure.
Ixabepilone Epothilone B analogue D (US) IV Promotes tubulin polymerisation and stabilises microtubular function, causing cell cycle arrest at G2/M phase and subsequently induces apoptosis Locally advanced or metastatic breast cancer. Myelosuppression, peripheral neuropathy, myocardial ischaemia (uncommon/rare), supraventricular arrhythmia (uncommon/rare) and hypersensitivity reaction (uncommon/rare).
Mitomycin Aziridine D (Au) IV Cross-links DNA Anal and bladder cancer Myelosuppression, pulmonary toxicity and haemolytic uraemic syndrome (rare).
Procarbazine Methylhydrazine D (Au) IM, IV Inhibits DNA, RNA and protein synthesis. Glioma and Hodgkin's lymphoma. Myelosuppression, neurotoxicity, pulmonary fibrosis (uncommon/rare), pneumonitis (uncommon/rare), haemolysis (uncommon/rare) and hepatic dysfunction (uncommon/rare).
2. Targeted antineoplastics
2.1 Monoclonal antibodies
Alemtuzumab Protein B2 (Au) IV CD52 antibody induces apoptosis in the tagged cells. Chronic lymphocytic leukaemia Pancytopenia, pneumonitis, arrhythmias and hypersensitivity reactions (rare), autoimmune haemolytic anaemia (rare), autoimmune thrombocytopenia (rare) and progressive multifocal leucoencephalopathy (rare).
Bevacizumab Protein D (Au) IV VEGF inhibitor. Colorectal, breast, ovarian, renal cell and non-squamous non-small cell lung cancer and glioblastoma Hypertension, thromboembolisms, heart failure, bleeding, neutropenia, thrombocytopenia, GI perforation, fistula formation, hypertensive encephalopathy, pulmonary hypertension, reversible posterior leucoencephalopathy syndrome, nasal septum perforation and osteonecrosis of the jaw.
Cetuximab Protein D (Au) IV EGFR inhibitor. Squamous cell head and neck cancer or EGFR-positive and KRAS wild-type metastatic colorectal cancer. Infusion-related reactions, skin reactions, hypomagnesaemia, hypocalcaemia, hypokalaemia, blood clots, interstitial lung disease and aseptic meningitis.
Denosumab Protein X (US) SC RANKL inhibitor. Osteoporosis, including drug and cancer related osteoporosis,giant cell tumour of bone and hypercalcaemia of malignancies Hypercholesterolaemia, cataract, urinary retention, hypocalcaemia, osteonecrosis of the jaw and anaphylaxis.
Gemtuzumab ozogamicin [162] Protein D (US) IV CD33 antibody that induces apoptosis of the tagged cell. Acute myeloid leukaemia Hepatic veno-occlusive disease, myelosuppression, cytokine release syndrome, hypersensitivity and electrolyte anomalies.
Ibritumomab tiuxetan [163][164] Protein D (US) IV CD20 antibody bound with the radioactive isotope, 90Y, induces radiation-dependent cell lysis. Non-Hodgkin's lymphoma and follicular lymphoma. Thrombocytopenia, neutropenia, anaemia, hypotension and secondary malignancies.
Ipilimumab Protein C (Au) IV CTLA4 antibody that causes immune system-mediated lysis of the tagged cell Unresectable or metastatic malignant melanoma. Life-threatening immune mediated reactions and fever.
Ofatumumab [165][166][167] Protein C (US) IV Anti-CD20 antibody. Chronic lymphocytic leukaemia Neutropenia, pneumonia, infusion reactions, cytopenias
Panitumumab Protein C (Au) IV EGFR inhibitor. RAS (KRAS or NRAS) wild-type metastatic colorectal cancer Skin reactions, electrolyte anomalies, anaphylaxis and angiooedema (rare).
Pertuzumab Protein D (Au) IV HER2 inhibitor. HER2-positive breast cancer. Anaphylaxis, cardiac dysfunction and anaemia.
Rituximab Protein C (Au) IV Anti-CD20 antibody. CD20-positive B cell non-Hodgkin lymphoma, chronic lymphocytic leukaemia, rheumatoid arthritis, Wegener's granulomatosis and microscopic polyangiitis Infusion-related reactions, neutropenia, arrhythmias, infection, thrombocytopenia (uncommon), anaemia (uncommon), angina (uncommon), myocardial infarction (uncommon), heart failure (uncommon), haemolytic anaemia (rare), aplastic anaemia (rare), serum sickness (rare), severe skin conditions (rare), pulmonary infiltrates (rare), pneumonitis (rare), cranial neuropathy (vision or hearing loss; rare) and progressive multifocal leucoencephalopathy (rare).
Tositumomab [168][169] Protein X (US) IV Anti-CD20 antibody which is tagged with I131. Non-Hodgkin's lymphoma Grade 3-4 cytopenias, methaemoglobinaemia, acute myeloid leukaemia or myelodysplastic syndrome, anaphylaxis and hyperthyroidism.
Trastuzumab Protein B2 (Au) IV Anti-HER2 antibody. HER2-positive breast cancer, gastric cancer, pancreatic cancer (orphan) and gastro-oesophageal junction cancer. Cardiac dysfunction, infusion-related reactions, peripheral neuropathy and pulmonary toxicity (rare).
2.2 Tyrosine kinase inhibitor
Afatinib [170] Phenyl quinazolinamine D (US) PO EGFR, HER2 and HER4 inhibitor. Non-small cell lung cancer. Diarrhoea, hypokalaemia, interstitial lung disease and hepatotoxicity.
Aflibercept [171] Protein C (US) IV VEGF and PGF inhibitor. Colorectal cancer. Myelosuppression, hypertension, dehydration, blood clots, GI perforation and reversible posterior leucoencephalopathy syndrome (uncommon).
Axitinib [172] Indazole & pyridine analogue D (Au) PO Multikinase inhibitor. Renal cell carcinoma Hypertension, thyroid dysfunction, blood clots, electrolyte disturbances, GI perforation (rare), fistula formation (rare), reversible posterior leucoencephalopathy syndrome (rare) and polycythaemia (uncommon).
Bosutinib [173][174] Quinoline carbonitrile analogue D (Au) PO Bcr-Abl and SRc kinase inhibitor. Chronic myeloid leukaemia Diarrhoea, thrombocytopenia, neutropenia, hepatotoxicity, QT interval prolongation, kidney failure, pleural effusion, pericarditis (uncommon/rare), acute pancreatitis (uncommon/rare), GI haemorrhage (uncommon/rare), anaphylactic shock (uncommon/rare), acute pulmonary oedema (uncommon/rare), respiratory failure (uncommon/rare), pulmonary hypertension(uncommon/rare) and erythema multiforme (uncommon/rare).
Crizotinib [175][176][177]
[178][179]
Aminopyridine D (US) PO ALK, Hepatocyte Growth Factor Receptor (HGFR, c-Met), and Recepteur d’Origine Nantais (RON) inhibitor. Non-small cell lung cancer Lymphopenia, neutropenia, hypophosphataemia, hypokalaemia, peripheral neuropathy, blood clots, QT interval prolongation, bradycardia, pneumonia, pneumonitis, kidney cyst, ARDS and liver failure.
Dasatinib Piperazine D (Au) PO BCR-ABL, SRC family, c-Kit, EPHA2 and PDGFR-β kinase inhibitor. Philadelphia positive chronic myeloid leukaemia and acute lymphoblastic leukaemia. Fluid retention, myelosuppression, haemorrhage, hypertension, electrolyte anomalies, cardiac dysfunction (rare), heart failure (rare), myocardial infarction, arrhythmia (rare), prolonged QT interval (rare), kidney failure (rare), hypersensitivity (rare) and hepatic failure (rare).
Erlotinib Quinazoline C (Au) PO EGFR inhibitor. Non-small cell lung cancer and pancreatic cancer. Skin reactions, diarrhoea, GI bleeds, anaemia, dehydration, interstitial lung disease (uncommon), hepatic failure (rare), hepatorenal syndrome (rare), GI perforation (rare) and ulcerative keratitis (rare).
Gefitinib Quinazoline C (Au) PO EGFR inhibitor. EGFR-mutation positive non-small cell lung cancer. Skin reactions, diarrhoea, dehydration, haemorrhage, interstitial lung disease (uncommon), pancreatitis (uncommon), hepatitis (uncommon), allergy (uncommon), hepatic failure (rare), toxic epidermal necrolysis (rare) and Stevens–Johnson syndrome (rare).
Imatinib Pyrimidine and piperazine. D (Au) PO Bcr-Abl kinase inhibitor. Philadelphia chromosome-positive acute lymphoblastic leukaemia, chronic myeloid leukaemia, GI stromal tumour and myelodysplastic/myeloproliferative diseases. Myelosuppression, fluid retention, GI bleeding, electrolyte anomalies, left ventricular dysfunction (uncommon), heart failure (uncommon), pulmonary oedema (uncommon), kidney failure (uncommon), angiooedema (rare), anaphylaxis (rare), GI perforation (rare), hepatotoxicity (rare), avascular necrosis (rare), myopathy (rare) and rhabdomyolysis (rare).
Lapatinib Quinazoline C (Au) PO HER2 inhibitor. HER2-positive breast cancer, stomach cancer (orphan) and oesophageal cancer (orphan). Diarrhoea, interstitial lung disease (uncommon), hepatotoxicity (uncommon) and anaphylaxis (rare).
Nilotinib Imidazole D (Au) PO Bcr-Abl kinase inhibitor. Chronic myeloid leukaemia. Myelosuppression, electrolyte disturbances, hyperglycaemia, prolonged QT interval (uncommon), peripheral arterial occlusive disease (uncommon), pancreatitis (uncommon), pleural effusion (uncommon) and pericardial effusion (uncommon).
Pazopanib Indazole D (Au) PO Multikinase inhibitor, including c-KIT, FGFR, PDGFR and VEGFR. Renal cell carcinoma and soft tissue sarcoma. Hypertension, QT interval prolongation, haemorrhage, blood clots, neutropenia, thrombocytopenia, neutropenia, thrombocytopenia, elevated thyroid-stimulating hormone, hypothyroidism, electrolyte disturbances, hypo- or hyperglycaemia, torsades de pointes (uncommon), heart failure (uncommon), hepatic failure (uncommon), GI perforation (uncommon), fistula formation (uncommon) and reversible posterior leucoencephalopathy syndrome (rare).
Ponatinib [180][181] Imidazopyridazine D (US) PO Multikinase inhibitor (BEGFR, PDGFR, FGFR, EPH receptors and SRC families of kinases, and KIT, RET, TIE2 and FLT3), that also inhibits T135I Bcr-Abl kinase. T135I positive Chronic myeloid leukaemia and Philadelphia chromosome positive acute lymphoblastic leukaemia. Hypertension, neutropenia, leucopenia, anaemia, thrombocytopenia, lymphopenia, pleural effusion, heart failure, peripheral neuropathy, haemorrhage, blood clots, pancreatitis and infection.
Regorafenib [182][183][184]
[185]
4-phenoxypyridine D (US) PO Multikinase inhibitor for RET, VEGFR1, VEGFR2, VEGFR3, KIT, PDGFR-alpha, PDGFR-beta, FGFR1, FGFR2, TIE2, DDR2, Trk2A, Eph2A, RAF-1, BRAF, BRAFV600E, SAPK2, PTK5, and Bcr-Abl. Colorectal cancer and GI stromal tumours. Anaemia, lymphopenia, thrombocytopenia, electrolyte anomalies, hepatotoxicity, hypertension, hypothyroidism, neutropenia, myocardial ischaemia or infarction.
Ruxolitinib [186][187][188]
[189]
Pyrimidinylpyrazole C (Au) PO JAK1 and JAK2 inhibitor. Myelofibrosis and pancreatic cancer (orphan). Anaemia and thrombocytopenia.
Sorafenib 4-phenoxypyridine D (Au) PO Multikinase inhibitor (including VEGF and PDGF receptor kinases). Renal cell carcinoma and hepatocellular carcinoma. Hypertension, skin reactions, bleeding, neutropenia, thrombocytopenia, lymphopenia, peripheral neuropathy, thyroid dysfunction, electrolyte anomalies, myocardial ischaemia or infarctions, heart failure (uncommon), GI perforation (uncommon), pancreatitis (uncommon), reversible posterior leucoencephalopathy syndrome (rare), hepatitis (rare), nephrotic syndrome (rare) and prolonged QT interval (rare).
Sunitinib Oxindole D (Au) PO Multikinase inhibitor (including VEGF & PDGF receptor tyrosine kinases) renal cell carcinoma, GI stromal tumour and pancreatic neuroendocrine tumour Neutropenia, thrombocytopenia, lymphopenia, hypertension, left ventricular dysfunction, heart failure, blood clots, thyroid dysfunction, electrolyte anomalies, pancreatitis (uncommon), hepatic failure (uncommon), prolonged QT interval (rare), torsades de pointes (rare), GI perforation (rare), fistula formation (rare), seizures (rare), reversible posterior leucoencephalopathy syndrome (rare), haemolytic uraemic syndrome (rare), thrombotic thrombocytopenic purpura (rare), nephrotic syndrome (rare), hypersensitivity (rare), angiooedema (rare), toxic epidermal necrolysis (rare) and Stevens–Johnson syndrome (rare).
Vandetanib[190] Quinazoline D (US) PO Tyrosine kinase inhibitor (TKI) with selective activity against RET, VEGFR-2 and EGFR Medullary thyroid cancer. Diarrhoea, hypertension, QT interval prolongation, depression, electrolyte anomalies, hypothyroidism and GI perforation (uncommon).
2.3 mTOR inhibitors
Everolimus Sirolimus analogue. C (Au) PO mTOR inhibitor. Renal cell cancer, pancreatic neuroendocrine tumour and breast cancer Pleural effusion, hyperglycaemia, hypercholesterolaemia, hypertriglyceridaemia, neutropenia, lymphopenia, thrombocytopenia, anaemia, bleeding, kidney failure, hypokalaemia, hypophosphataemia, pneumonitis, impaired wound healing (uncommon), anaphylaxis (rare) and angiooedema (rare).
Temsirolimus Sirolimus analogue. D (Au) IV mTOR inhibitor. Renal cell cancer and mantle cell lymphoma. Infusion reactions, impaired wound healing, hyperglycaemia, hypercholesterolaemia, hypertriglyceridaemia, neutropenia, lymphopenia, thrombocytopenia, anaemia, bleeding, kidney failure, hypokalaemia, hypophosphataemia, pneumonitis, bowel perforation (uncommon) and intracerebral bleeding and Stevens–Johnson syndrome (rare).
2.4 Retinoids
Alitretinoin 9-cis retinoic acid D (US) Topical Retinoic acid receptor (RAR) and retinoid X receptor (RXR) agonist. Kaposi's sarcoma. Oedema, rashes
Bexarotene[191] Phenethylnaphthalene X (US) PO, topical RXR agonist. Cutaneous T cell lymphoma Leucopenia, anaemia, lactic dehydrogenase increased, hypochromic anaemia, hyperlipidaemia, hypercholesteraemia, hypothyroidism, haemorrhage, hypertension and kidney dysfunction.
Isotretinoin 13-cis retinoic acid D (Au, topical), X (Au, oral) PO, topical RXR & RAR agonist. Neuroblastoma[192] andacne. Topical: Skin reactions, blood lipid anomalies, increased platelet count and osteoporosis.[193] Oral: Anaemia, Red blood cell sedimentation rate increased, thrombocytopenia, thrombocytosis, neutropenia, anaphylaxis, hypersensitivity, diabetes mellitus, hyperuricaemia, psychiatric disturbances (rare), convulsions (very rare), conjunctivitis, vasculitis (very rare), GI haemorrhage (very rare), hepatitis (very rare), erythema multiforme, Stevens–Johnson Syndrome, toxic epidermal necrolysis, arthritis (very rare), rhabdomyolysis and glomerulonephritis (very rare).[194]
Tamibarotene [195][196][197] Naphthalene N/A PO RAR agonist. Refractory acute promyelocytic leukaemia and Alzheimer's disease. Hypercholesterolaemia, hypertriglyceridaemia, gastrointestinal disturbances, liver damage, leucocytosis anddifferentiation syndrome.
Tretinoin all-trans retinoic acid D (Au, topical), X (Au, oral) PO, topical RXR & RAR agonist. Acne andacute promyelocytic leukaemia. Oral: Differentiation syndrome, hyperleucocytosis, elevated cholesterol and triglycerides, arrhythmias, pancreatitis, elevated liver enzymes, thrombosis, intracranial hypertension and pseudotumour cerebri (mainly in children), anxiety, depression and genital ulceration (rare). Topical: Erythema.
2.4 Immunomodulatory Agents (IMiDs)
Lenalidomide Thalidomide analogue X (Au) PO Numerous actions; anti-angiogenesis (via inhibition of VEGF release), anti-TNF, IL-6 and pro-IL-2, IFN-γ effects. Also stimulates T cells and apoptosis in cancer cells. Multiple myeloma Blood clots, neutropenia (dose-limiting), thrombocytopenia (dose-limiting), anaemia, infection, hypotension, hypokalaemia, hypothyroidism, Stevens–Johnson syndrome, toxic epidermal necrolysis, angioedema, pneumonitis, hepatotoxicity and secondary malignancies (mostly myelodysplastic syndrome and acute myeloid leukaemia).
Pomalidomide Thalidomide analogue X (US) PO As above. Multiple myeloma and systemic sclerosis (orphan). Neutropenia, anaemia, pneumonia, thrombocytopenia, hypercalcaemia, hyperglycaemia, kidney failure, lymphopenia, hyponatraemia, hypocalcaemia, hypokalaemia, peripheral neuropathy and thromboembolism.
Thalidomide Thalidomide X (Au) PO As above. Multiple myeloma, erythema nodosum leprosum and the following orphan indications: graft versus host disease, mycobacterial infection, recurrent aphthous ulcers, severe recurrent aphthous stomatitis, primary brain malignancies, HIV-associated wasting syndrome, Crohn's disease, Kaposi's sarcoma, myelodysplastic syndrome and haematopoietic stem cell transplantation. Peripheral neuropathy, depression, thromboembolism, bradycardia, orthostatic hypotension, leucopenia, hypothyroidism, thrombocytopenia (uncommon), Stevens–Johnson syndrome (rare), toxic epidermal necrolysis (rare), pneumonitis (rare), hepatotoxicity (rare) and hearing loss (rare).
2.5 Histone deacetylase inhibitors
Romidepsin [198] Depsipeptide D (US) IV Histone deacetylase inhibitor, hence inducing alterations in gene expression in the affected cells. Peripheral and cutaneous T cell lymphoma. Electrolyte anomalies, anaemia, thrombocytopenia, neutropenia, lymphopenia and ECG anomalies.
Valproate[Note 1] [199][200][201]
[202]
Branched short-chain fatty acids. D (Au) PO, IV As above. Migraine prophylaxis, mania, epilepsy, fragile X syndrome (orphan), familial adenomatous polyposis (orphan) and the following off-label uses: cervical cancer, melanoma, mesothelioma, acute myeloid leukaemia and myelodysplastic syndrome. Hyperammonaemia, thrombocytopenia, polycystic ovaries, SIADH (uncommon), hepatic failure (rare), pancreatitis (rare), leucopenia (rare), neutropenia (rare), pure red cell aplasia (rare), agranulocytosis (rare), extrapyramidal syndrome (rare), reduced BMD with long-term use, pleural effusion (rare) and multiorgan hypersensitivity reaction (rare).
Vorinostat [203] Phenyloctanediamide D (US) PO As above. As per romidepsin. Thrombocytopenia, anaemia, QT interval prolongation and pulmonary embolism.
2.6 Other Agents
Anagrelide Imidazoquinazoline B3 (Au) PO Phosphodiesterase 3 inhibitor. Essential thrombocythaemia Fluid retention, palpitations, tachycardia, hepatotoxicity (uncommon), heart failure (uncommon), hypertension (uncommon), arrhythmia (uncommon), syncope (uncommon), cardiomyopathy (rare), cardiomegaly (rare), MI (rare), pulmonary hypertension (rare), interstitial lung disease (rare) and pancreatitis (rare).
Arsenic trioxide[Note 2] Arsenic compound X (Au) IV Not fully understood. Induces partial differentiation and promotes apoptosis of leukaemic cells and may also inhibit angiogenesis. Refractory or relapsed acute promyelocytic leukaemia. Orphan indications include: acute myeloid leukaemia, chronic lymphocytic leukaemia, malignant glioma, myelodysplastic syndrome, multiple myeloma, liver cancer and chronic myeloid leukaemia. Differentiation syndrome, hyperleucocytosis, neutropenia, thrombocytopenia, ventricular tachycardia, prolonged QT interval, torsades de pointes, complete atrioventricular block, peripheral neuropathy, hyperglycaemia, hypokalaemia, hypomagnesaemia, elevation of bilirubin or aminotransferases, hepatotoxicity and secondary malignancies.
Asparaginase
[Note 3]
Protein D (Au) IM, IV Catalyses the conversion of the amino acid L-asparagine to aspartic acid and thereby reduces the availability of L-asparagine to leukaemic cells. Unlike normal cells, certain types of leukaemic cells do not synthesise L-asparagine, which is essential for cell growth and survival. Acute lymphoblastic leukaemia and lymphoblastic lymphoma. Allergic reactions, haemorrhagic and thrombotic events, uraemia, pancreatitis, hyperglycaemia, hyperammonaemia, acute kidney failure and diabetic ketoacidosis.
BCG vaccine
[Note 4]
Protein B2 (Au) IB Live, attenuated Mycobacterium bovis, which produces a local inflammatory reaction, resulting in elimination or reduction of superficial tumour lesions of the bladder. Bladder cancer Cystitis, BCG infection and contracted bladder.
Denileukin diftitox [204][205][206]
[207]
Protein C (US) IV Interleukin 2 combined with diphtheria toxin which binds to the interleukin receptor on immune cells and introduces the diphtheria toxin into the cell. Cutaneous T cell lymphoma and peripheral T cell lymphoma (orphan). Infusion reactions, hypocalcaemia, hypotension, thrombocytopenia, Acute renal insufficiency (uncommon/rare), Hyper/hypothyroidism (uncommon/rare), pancreatitis (uncommon/rare) and toxic epidermal necrolysis (uncommon/rare).
Vemurafenib Pyrrolopyridine D (Au) PO BRAF kinase inhibitor. BRAF kinase mutation V600E-positive Metastatic melanoma. Skin reactions, secondary malignancies (mostly squamous cell carcinoma), anaphylaxis (rare) and hypotension (rare).

Abbreviations/Acronyms:
IM - Intramuscular. IV - Intravenous. IA - Intra-arterial. SC - Subcutaneous. PO - Per os, oral. IP - Intrapleural. IB - Intrabladder. Preg. cat. - Pregnancy category. The preferred pregnancy category is Australian, but if it is unavailable the pregnancy category given is American.

Notes edit

  1. ^ Its use in cancer treatment is purely investigational at this point in time
  2. ^ There is no INN for arsenic trioxide, just the USAN
  3. ^ There is no INN for asparaginase, only a USAN
  4. ^ There is no INN for BCG

Reference list edit

  1. ^ a b c d e f g Rossi, S, ed. (2013). Australian Medicines Handbook (2013 ed.). Adelaide: The Australian Medicines Handbook Unit Trust. ISBN 978-0-9805790-9-3.
  2. ^ a b c Joint Formulary Committee (2013). British National Formulary (BNF) (65 ed.). London, UK: Pharmaceutical Press. ISBN 978-0-85711-084-8. {{cite book}}: Cite has empty unknown parameter: |coauthors= (help)
  3. ^ Brunton, L; Chabner, B; Knollman, B (2010). Goodman and Gilman's The Pharmacological Basis of Therapeutics (12th ed.). New York: McGraw-Hill Professional. ISBN 978-0-07-162442-8.{{cite book}}: CS1 maint: multiple names: authors list (link)
  4. ^ a b c Alldredge, BK; Corelli, RL; Ernst, ME; Guglielmo, BJ; Jacobson, PA; Kradjan, WA; Williams, BR (February 2012). Applied therapeutics: the clinical use of drugs. Koda Kimble and Youngs Applied Therapeutics (10th ed.). Baltimore, MD: Lippincott Williams & Wilkins. ISBN 978-1609137137.{{cite book}}: CS1 maint: multiple names: authors list (link)
  5. ^ Sweetman, S (ed.). Martindale: The Complete Drug Reference. London, UK: Pharmaceutical Press. Retrieved 8 February 2014.
  6. ^ a b Momparler, RL (August 2012). "A Perspective on the Comparative Antileukemic Activity of 5-Aza-2'-deoxycytidine (Decitabine) and 5-Azacytidine (Vidaza)" (PDF). Pharmaceuticals (Basel, Switzerland). 5 (8): 875–881. doi:10.3390/ph5080875. PMC 3763670. PMID 24280679.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  7. ^ a b Estey, EH (12 June 2013). "Epigenetics in clinical practice: the examples of azacitidine and decitabine in myelodysplasia and acute myeloid leukemia". Leukemia. 27 (9): 1803–1812. doi:10.1038/leu.2013.173. PMID 23757301.
  8. ^ Kim, YJ (June 2013). "Use of azacitidine for myelodysplastic syndromes: controversial issues and practical recommendations" (PDF). Blood research. 48 (2): 87–98. doi:10.5045/br.2013.48.2.87. PMC 3698413. PMID 23826577. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  9. ^ Platzbecker, U (September 2013). "Combination of azacitidine and lenalidomide in myelodysplastic syndromes or acute myeloid leukemia-a wise liaison?". Leukemia. 27 (9): 1813–9. doi:10.1038/leu.2013.140. PMID 23644421. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  10. ^ Ritchie, EK (2012). "Safety and efficacy of azacitidine in the treatment of elderly patients with myelodysplastic syndrome" (PDF). Clinical interventions in aging. 7: 165–73. doi:10.2147/CIA.S24659. PMC 3393359. PMID 22791989.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  11. ^ Keating, GM (28 May 2012). "Azacitidine: a review of its use in the management of myelodysplastic syndromes/acute myeloid leukaemia". Drugs. 72 (8): 1111–36. doi:10.2165/11209430-000000000-00000. PMID 22571445.
  12. ^ Martínez-Francés, A (June 2011). "Adverse effects of azacitidine: onset, duration, and treatment". Advances in therapy. 28 Suppl 4: 1–5. doi:10.1007/s12325-011-0021-5. PMID 21688206.
  13. ^ Font, P (March 2011). "Azacitidine for the treatment of patients with acute myeloid leukemia with 20%-30% blasts and multilineage dysplasia". Advances in therapy. 28 Suppl 3: 1–9. doi:10.1007/s12325-011-0002-8. PMID 21431628.
  14. ^ Vigil, CE (24 September 2010). "Safety and efficacy of azacitidine in myelodysplastic syndromes" (PDF). Drug design, development and therapy. 4: 221–9. doi:10.2147/DDDT.S3143. PMC 2948932. PMID 20957213. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  15. ^ McCormack, SE (7 September 2010). "Epigenetic approaches in the treatment of myelodysplastic syndromes: clinical utility of azacitidine" (PDF). OncoTargets and therapy. 3: 157–65. doi:10.2147/OTT.S5852. PMC 2939768. PMID 20856790. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  16. ^ Martens, UM, ed. (2010). "11 5-Azacytidine/Azacitidine". Small molecules in oncology. Recent Results in Cancer Research. Vol. 184. Heidelberg: Springer. p. 159-170. doi:10.1007/978-3-642-01222-8. ISBN 978-3-642-01222-8.
  17. ^ Chintala, L (9 April 2011). "Capecitabine versus 5-fluorouracil in colorectal cancer: where are we now?" (PDF). Oncology Reviews. 5 (2): 129–140. doi:10.1007/s12156-011-0074-3. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  18. ^ Bang, YJ (December 2011). "Capecitabine in gastric cancer". Expert review of anticancer therapy. 11 (12): 1791–1806. doi:10.1586/era.11.172. PMID 22117147.
  19. ^ Hirsch, BR (2011). "Capecitabine in the management of colorectal cancer" (PDF). Cancer management and research. 3: 79–89. doi:10.2147/CMR.S11250. PMC 3097797. PMID 21629830. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  20. ^ Quidde, J (2012). "Clinical management of localized colon cancer with capecitabine" (PDF). Clinical Medicine Insights. Oncology. 6: 363–73. doi:10.4137/CMO.S8194. PMC 3498969. PMID 23170068. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  21. ^ Fernández-Martos, C (28 May 2012). "The role of capecitabine in locally advanced rectal cancer treatment: an update". Drugs. 72 (8): 1057–1073. doi:10.2165/11633870-000000000-00000. PMID 22621694. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  22. ^ Ma, Y (June 2012). "Capecitabine for the treatment for advanced gastric cancer: efficacy, safety and ethnicity". Journal of clinical pharmacy and therapeutics. 37 (3): 266–275. doi:10.1111/j.1365-2710.2011.01289.x. PMID 21950464. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  23. ^ O'Shaughnessy, JA (2012). "Capecitabine monotherapy: review of studies in first-line HER-2-negative metastatic breast cancer". The oncologist. 17 (4): 476–84. doi:10.1634/theoncologist.2011-0281. PMID 22418569. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  24. ^ Petrelli, F (June 2012). "5-Fluorouracil or capecitabine in the treatment of advanced colorectal cancer: a pooled-analysis of randomized trials". Medical oncology (Northwood, London, England). 29 (2): 1020–1029. doi:10.1007/s12032-011-9958-0. PMID 21516482. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  25. ^ Barrios, CH (26 January 2012). "The Role of Capecitabine in Early Stage Breast Cancer". Current Breast Cancer Reports. 4 (1): 22–29. doi:10.1007/s12609-011-0067-z.
  26. ^ Comi, G (January 2013). "Cladribine tablets for the treatment of relapsing-remitting multiple sclerosis". Expert opinion on pharmacotherapy. 14 (1): 123–36. doi:10.1517/14656566.2013.754012. PMID 23256518. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  27. ^ Spurgeon, S (August 2009). "Cladribine: not just another purine analogue?". Expert opinion on investigational drugs. 18 (8): 1169–81. doi:10.1517/13543780903071038. PMID 19604118. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  28. ^ Bagnato, F (October 2011). "Novel Agents and Emerging Treatment Strategies in Multiple Sclerosis. What Role for Cladribine?". Clinical Medicine Insights: Therapeutics: 425. doi:10.4137/CMT.S6456. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  29. ^ Leist, TP (January–February 2011). "Cladribine: mode of action and implications for treatment of multiple sclerosis". Clinical neuropharmacology. 34 (1): 28–35. doi:10.1097/WNF.0b013e318204cd90. PMID 21242742. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: date format (link)
  30. ^ Warnke, C (21 July 2010). "Identification of targets and new developments in the treatment of multiple sclerosis--focus on cladribine" (PDF). Drug design, development and therapy. 4: 117–26. doi:10.2147/DDDT.S6627. PMC 2915536. PMID 20689698. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  31. ^ Sigal, DS (21 October 2010). "Beyond hairy cell: the activity of cladribine in other hematologic malignancies" (PDF). Blood. 116 (16): 2884–96. doi:10.1182/blood-2010-02-246140. PMID 20634380. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  32. ^ Sipe, JC (March 2010). "Cladribine tablets: a potential new short-course annual treatment for relapsing multiple sclerosis". Expert review of neurotherapeutics. 10 (3): 365–75. doi:10.1586/ern.10.12. PMID 20187859.
  33. ^ Hartung, HP (February 2010). "Development of oral cladribine for the treatment of multiple sclerosis". Journal of neurology. 257 (2): 163–70. doi:10.1007/s00415-009-5359-0. PMID 19921304. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  34. ^ Warnke, C (January 2012). "Cladribine as a therapeutic option in multiple sclerosis". Clinical immunology (Orlando, Fla.). 142 (1): 68–75. doi:10.1016/j.clim.2011.05.009. PMID 21733757. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  35. ^ Warnke, C (21 July 2010). "Identification of targets and new developments in the treatment of multiple sclerosis--focus on cladribine" (PDF). Drug design, development and therapy. 4: 117–126. doi:10.2147/DDDT.S6627. PMC 2915536. PMID 20689698. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  36. ^ Bryson, HM (November 1993). "Cladribine. A review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in haematological malignancies". Drugs. 46 (5): 872–894. doi:10.2165/00003495-199346050-00007. PMID 7507037. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  37. ^ Robak, T (January 2006). "Older and new formulations of cladribine. Pharmacology and clinical efficacy in hematological malignancies". Recent patents on anti-cancer drug discovery. 1 (1): 23–38. doi:10.2174/157489206775246467. PMID 18221024. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  38. ^ Spurgeon, S (August 2009). "Cladribine: not just another purine analogue?". Expert opinion on investigational drugs. 18 (8): 1169–81. doi:10.1517/13543780903071038. PMID 19604118. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  39. ^ Hentosh, P (June 2010). "The cladribine conundrum: deciphering the drug's mechanism of action". Expert opinion on drug metabolism & toxicology. 6 (1): 75–81. doi:10.1517/17425250903393745. PMID 19968576. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  40. ^ Warnke, C (January 2012). "Cladribine as a therapeutic option in multiple sclerosis". Clinical Immunology. 142 (1): 68–75. doi:10.1016/j.clim.2011.05.009. PMID 21733757. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  41. ^ Kantarjian, HM (October 2007). "Clofarabine: past, present, and future". Leukemia & lymphoma. 48 (10): 1922–30. doi:10.1080/10428190701545644. PMID 17852710. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  42. ^ Harned, TM (April 2008). "Treating refractory leukemias in childhood, role of clofarabine" (PDF). Therapeutics and clinical risk management. 4 (2): 327–36. doi:10.2147/TCRM.S2941. PMC 2504075. PMID 18728851. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  43. ^ Thomas, X (October 2009). "Clofarabine for the treatment of adult acute myeloid leukemia". Future oncology (London, England). 5 (8): 1197–210. doi:10.2217/fon.09.105. PMID 19852733. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  44. ^ Lech-Maranda, E (June 2009). "Clofarabine as a novel nucleoside analogue approved to treat patients with haematological malignancies: mechanism of action and clinical activity". Mini reviews in medicinal chemistry. 9 (7): 805–812. doi:10.2174/138955709788452586. PMID 19519505. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  45. ^ Jeha, S (October 2009). "Recent progress in the treatment of acute lymphoblastic leukemia: clofarabine". Hematology/oncology clinics of North America. 23 (5): 1137–44, viii. doi:10.1016/j.hoc.2009.07.011. PMID 19825457.
  46. ^ Sampat, K (October 2009). "Clofarabine: emerging role in leukemias". Expert opinion on investigational drugs. 18 (10): 1559–1564. doi:10.1517/13543780903173222. PMID 19715446. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  47. ^ Larson, ML (June 2009). "Clofarabine: a new treatment option for patients with acute myeloid leukemia". Expert opinion on pharmacotherapy. 10 (8): 1353–1357. doi:10.1517/14656560902997990. PMID 19463072. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  48. ^ Ghanem, H (February 2010). "Clofarabine in leukemia". Expert review of hematology. 3 (1): 15–22. doi:10.1586/ehm.09.70. PMID 21082931. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  49. ^ Ghanem, H (April 2013). "The role of clofarabine in acute myeloid leukemia". Leukemia & lymphoma. 54 (4): 688–698. doi:10.3109/10428194.2012.726722. ISBN 978-0-12-374340-4. PMID 22957815. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  50. ^ Ghanem, H (April 2013). "The role of clofarabine in acute myeloid leukemia". Leukemia & Lymphoma. 54 (4): 688–698. doi:10.3109/10428194.2012.726722. PMID 22957815. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  51. ^ Hamada, A (2002). "Clinical pharmacokinetics of cytarabine formulations". Clinical pharmacokinetics. 41 (10): 705–718. doi:10.2165/00003088-200241100-00002. PMID 12162758. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  52. ^ Cros, E (June 2004). "Problems related to resistance to cytarabine in acute myeloid leukemia". Leukemia & lymphoma. 45 (6): 1123–1132. doi:10.1080/1042819032000159861. PMID 15359991. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  53. ^ El‐Subbagh, HI (2009). "2 Cytarabine". In Brittain, HG (ed.). Profiles of drug substances, excipients and related methodology (PDF). London: Academic. pp. 37–113. doi:10.1016/S0099-5428(08)00002-6. ISBN 978-0-12-374340-4. ISSN 0099-5428. {{cite book}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  54. ^ "Dacogen (decitabine) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 10 February 2014.
  55. ^ Plimack, ER (January 2007). "Decitabine and its role in the treatment of hematopoietic malignancies". Leukemia & Lymphoma. 48 (8): 1472–1481. doi:10.1080/10428190701471981. PMID 7701577. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  56. ^ Oki, Y (February 2007). "Decitabine—Bedside to bench". Critical Reviews in Oncology/Hematology. 61 (2): 140–152. doi:10.1016/j.critrevonc.2006.07.010. PMID 17023173. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  57. ^ Jabbour, E (June 2008). "Evolution of decitabine development: accomplishments, ongoing investigations, and future strategies" (PDF). Cancer. 112 (11): 2341–51. doi:10.1002/cncr.23463. PMID 18398832. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  58. ^ Santos, FP (January 2010). "Decitabine in the treatment of myelodysplastic syndromes" (PDF). Expert review of anticancer therapy. 10 (1): 9–22. doi:10.1586/era.09.164. PMC 2376088. PMID 20014881. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  59. ^ Daskalakis, M (2010). "Decitabine". Recent results in cancer research. Fortschritte der Krebsforschung. Progres dans les recherches sur le cancer. 184: 131–57. doi:10.1007/978-3-642-01222-8_10. PMID 20072836. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  60. ^ Marks, PW (March 2012). "Decitabine for acute myeloid leukemia". Expert review of anticancer therapy. 12 (3): 299–305. doi:10.1586/era.11.207. PMID 22369322.
  61. ^ Garcia, JS (June 2010). "An update on the safety and efficacy of decitabine in the treatment of myelodysplastic syndromes" (PDF). OncoTargets and therapy. 3: 1–13. doi:10.2147/OTT.S7222. PMC 2895778. PMID 20616953. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  62. ^ Curran, MP (12 April 2013). "Decitabine: A Review of its Use in Older Patients with Acute Myeloid Leukaemia". Drugs & Aging. 30 (6): 447–458. doi:10.1007/s40266-013-0084-x. PMID 23580320.
  63. ^ "FUDR (floxuridine) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 10 February 2014.
  64. ^ Montillo, M (September 2006). "Role of fludarabine in hematological malignancies". Expert review of anticancer therapy. 6 (9): 1141–61. doi:10.1586/14737140.6.9.1141. PMID 17020450. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  65. ^ Anderson, VR (2007). "Fludarabine: a review of its use in non-Hodgkin's lymphoma". Drugs. 67 (11): 1633–55. doi:10.2165/00003495-200767110-00008. PMID 17661532. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  66. ^ Ricci, F (February 2009). "Fludarabine in the treatment of chronic lymphocytic leukemia: a review" (PDF). Therapeutics and clinical risk management. 5 (1): 187–207. doi:10.2147/TCRM.S3688. PMC 2697528. PMID 19436622. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  67. ^ Ricci, F (February 2009). "Fludarabine in the treatment of chronic lymphocytic leukemia: a review" (PDF). Therapeutics and clinical risk management. 5 (1): 187–207. doi:10.2147/TCRM.S3688. PMC 2697528. PMID 19436622. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  68. ^ Souchet-Compain, L (June 2013). "Fludarabine in Waldenstrom's macroglobulinemia". Expert review of hematology. 6 (3): 229–37. doi:10.1586/ehm.13.17. PMID 23782076. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  69. ^ Plosker, GL (2003). "Oral fludarabine". Drugs. 63 (21): 2317–23. doi:10.2165/00003495-200363210-00004. PMID 14524733. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  70. ^ Lukenbill, J (September 2013). "Fludarabine: a review of the clear benefits and potential harms". Leukemia research. 37 (9): 986–94. doi:10.1016/j.leukres.2013.05.004. PMID 23787174. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  71. ^ Grem, JL (November 2000). "5-Fluorouracil: forty-plus and still ticking. A review of its preclinical and clinical development". Investigational new drugs. 18 (4): 299–313. doi:10.1023/A:1006416410198. PMID 11081567.
  72. ^ Abraham, LM (2007). "The clinical applications of fluorouracil in ophthalmic practice". Drugs. 67 (2): 237–55. doi:10.2165/00003495-200767020-00005. PMID 17284086. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  73. ^ Patel, PA (April 2011). "Evolution of 5-fluorouracil-based chemoradiation in the management of rectal cancer". Anti-cancer drugs. 22 (4): 311–6. doi:10.1097/CAD.0b013e3283441a63. PMID 21301320.
  74. ^ Noble, S (September 1997). "Gemcitabine. A review of its pharmacology and clinical potential in non-small cell lung cancer and pancreatic cancer". Drugs. 54 (3): 447–472. doi:10.2165/00003495-199754030-00009. PMID 9279506. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  75. ^ Aapro, MS (March 1998). "Gemcitabine--a safety review". Anti-cancer drugs. 9 (3): 191–201. PMID 9625429. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  76. ^ Ryan, CW (March 2000). "Gemcitabine in the treatment of bladder cancer". Expert opinion on pharmacotherapy. 1 (3): 547–553. PMID 11249537. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  77. ^ Manegold, C (February 2000). "Gemcitabine in non-small cell lung cancer (NSCLC)". Investigational new drugs. 18 (1): 29–42. doi:10.1023/A:1006327729228. PMID 10830139. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  78. ^ Oettle, H (November 2000). "The role of gemcitabine alone and in combination in the treatment of pancreatic cancer". Anti-cancer drugs. 11 (10): 771–86. doi:10.1097/00001813-200011000-00001. PMID 11142685. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  79. ^ Crinò, L (June 2002). "Gemcitabine in non-small cell lung cancer". Expert opinion on pharmacotherapy. 3 (6): 745–753. doi:10.1517/14656566.3.6.745. PMID 12036414. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  80. ^ Wong, A (2009). "Clinical pharmacology and pharmacogenetics of gemcitabine". Drug metabolism reviews. 41 (2): 77–88. doi:10.1080/03602530902741828. PMID 19514966. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  81. ^ Lennard, L (October 1992). "The clinical pharmacology of 6-mercaptopurine". European Journal of Clinical Pharmacology. 43 (4): 329–339. doi:10.1007/BF02220605. PMID 1451710.
  82. ^ Bradford, K (7 October 2011). "Optimizing 6-mercaptopurine and azathioprine therapy in the management of inflammatory bowel disease". World journal of gastroenterology. 17 (37): 4166–73. doi:10.3748/wjg.v17.i37.4166. PMID 22072847. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  83. ^ Timmer, A (12 September 2012). "Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis". The Cochrane database of systematic reviews. 9: CD000478. doi:10.1002/14651858.CD000478.pub3. PMID 22972046. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  84. ^ "Arranon (nelarabine) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 11 February 2014.
  85. ^ Reilly, KM (February 2009). "Profile of nelarabine: use in the treatment of T-cell acute lymphoblastic leukemia" (PDF). OncoTargets and therapy. 2: 219–28. PMC 2886323. PMID 20616909. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  86. ^ DeAngelo, DJ (October 2009). "Nelarabine for the treatment of patients with relapsed or refractory T-cell acute lymphoblastic leukemia or lymphoblastic lymphoma". Hematology/oncology clinics of North America. 23 (5): 1121–35, vii–viii. doi:10.1016/j.hoc.2009.07.008. PMID 19825456.
  87. ^ Roecker, AM (December 2010). "Nelarabine in the treatment of refractory T-cell malignancies" (PDF). Clinical Medicine Insights. Oncology. 4: 133–41. doi:10.4137/CMO.S4364. PMC 2999959. PMID 21151585. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  88. ^ Sanford, M (2008). "Nelarabine". Drugs. 68 (4): 439–47. doi:10.2165/00003495-200868040-00004. PMID 18318562. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  89. ^ Gandhi, V (November 2006). "Clofarabine and nelarabine: two new purine nucleoside analogs". Current opinion in oncology. 18 (6): 584–90. doi:10.1097/01.cco.0000245326.65152.af. PMID 16988579. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  90. ^ Curbo, S (September 2006). "Nelarabine: a new purine analog in the treatment of hematologic malignancies". Reviews on recent clinical trials. 1 (3): 185–92. doi:10.2174/157488706778250104. PMID 18473971. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  91. ^ Kline, J (September 2006). "Nelarabine in the treatment of refractory T-cell malignant diseases". Expert opinion on pharmacotherapy. 7 (13): 1791–9. doi:10.1517/14656566.7.13.1791. PMID 16925505. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  92. ^ Grever, MR (March 2003). "Pentostatin in the treatment of hairy-cell leukemia". Best practice & research. Clinical haematology. 16 (1): 91–9. doi:10.1016/S1521-6926(03)00002-1. PMID 12670468. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  93. ^ Higman, M (December 2004). "Pentostatin –pharmacology, immunology, and clinical effects in graft-versus-host disease". Expert Opinion on Pharmacotherapy. 5 (12): 2605–2613. doi:10.1517/14656566.5.12.2605. PMID 15571477. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  94. ^ Ho, AD (April 2006). "Pentostatin for the treatment of indolent lymphoproliferative disorders". Seminars in hematology. 43 (2 Suppl 2): S2-10. doi:10.1053/j.seminhematol.2005.12.005. PMID 16549110. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  95. ^ Grever, MR (October 2006). "Pentostatin: Impact on Outcome in Hairy Cell Leukemia". Hematology/Oncology Clinics of North America. 20 (5): 1099–1108. doi:10.1016/j.hoc.2006.06.001. PMID 16990110.
  96. ^ Sauter, C (September 2008). "Pentostatin in chronic lymphocytic leukemia". Expert opinion on drug metabolism & toxicology. 4 (9): 1217–22. doi:10.1517/17425255.4.9.1217. PMID 18721115. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  97. ^ Lamanna, N (June 2009). "Pentostatin treatment combinations in chronic lymphocytic leukemia". Clinical advances in hematology & oncology. 7 (6): 386–392. PMID 19606074. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  98. ^ Wellington, K (2001). "Oral tegafur/uracil". Drugs & aging. 18 (12): 935–48, discussion 949-50. PMID 11888348. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  99. ^ Takiuchi, H (August 1998). "Uracil-tegafur in gastric carcinoma: a comprehensive review". Journal of Clinical Oncology. 16 (8): 2877–85. PMID 9704742. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  100. ^ Ward, SE (3 July 2006). "The clinical and economic benefits of capecitabine and tegafur with uracil in metastatic colorectal cancer". British Journal of Cancer. 95 (1): 27–34. doi:10.1038/sj.bjc.6603215. PMID 16804526. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  101. ^ Casado, E (August 2008). "UFT (tegafur-uracil) in rectal cancer". Annals of Oncology. 19 (8): 1371–8. doi:10.1093/annonc/mdn067. PMID 18381370. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  102. ^ Ishikawa, T (14 May 2008). "Chemotherapy with enteric-coated tegafur/uracil for advanced hepatocellular carcinoma". World journal of gastroenterology. 14 (18): 2797–801. doi:10.3748/wjg.14.2797. PMID 18473401.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  103. ^ Oba, K (April 2009). "Efficacy of adjuvant chemotherapy using tegafur-based regimen for curatively resected gastric cancer: update of a meta-analysis". International journal of clinical oncology. 14 (2): 85–9. doi:10.1007/s10147-009-0877-4. PMID 19390937.
  104. ^ Matt, P (2011). "The European Medicines Agency review of Tegafur/Gimeracil/Oteracil (Teysuno™) for the treatment of advanced gastric cancer when given in combination with cisplatin: summary of the Scientific Assessment of the Committee for medicinal products for human use (CHMP)". The oncologist. 16 (10): 1451–7. doi:10.1634/theoncologist.2011-0224. PMID 21963999. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  105. ^ Estlin, EJ (December 2001). "Continuing therapy for childhood acute lymphoblastic leukaemia: clinical and cellular pharmacology of methotrexate, 6-mercaptopurine and 6-thioguanine". Cancer treatment reviews. 27 (6): 351–63. doi:10.1053/ctrv.2002.0245. PMID 11908928.
  106. ^ Elgemeie, GH (2003). "Thioguanine, mercaptopurine: their analogs and nucleosides as antimetabolites". Current pharmaceutical design. 9 (31): 2627–42. doi:10.2174/1381612033453677. PMID 14529546.
  107. ^ Duley, JA (October 2005). "Thiopurine therapies: problems, complexities, and progress with monitoring thioguanine nucleotides". Therapeutic drug monitoring. 27 (5): 647–654. doi:10.1097/01.ftd.0000169061.52715.3e. PMID 16175140. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  108. ^ De Bruyne, R (February 2006). "Chronic liver disease related to 6-thioguanine in children with acute lymphoblastic leukaemia". Journal of hepatology. 44 (2): 407–10. doi:10.1016/j.jhep.2005.06.020. PMID 16226335. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  109. ^ Seinen, ML (September 2010). "Dosing 6-thioguanine in inflammatory bowel disease: expert-based guidelines for daily practice" (PDF). Journal of gastrointestinal and liver diseases. 19 (3): 291–4. PMID 20922194. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  110. ^ Tung, JP (November 1990). "The practical use of methotrexate in psoriasis". Drugs. 40 (5): 697–712. doi:10.2165/00003495-199040050-00005. PMID 2292232. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  111. ^ Bannwarth, B (January 1994). "Methotrexate in rheumatoid arthritis. An update". Drugs. 47 (1): 25–50. doi:10.2165/00003495-199447010-00003. PMID 7510620. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  112. ^ Barnhart, K (March 2001). "The pharmacology of methotrexate". Expert opinion on pharmacotherapy. 2 (3): 409–17. doi:10.1517/14656566.2.3.409. PMID 11336595. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  113. ^ Khan, ZA (February 2012). "Methotrexate: a detailed review on drug delivery and clinical aspects". Expert opinion on drug delivery. 9 (2): 151–69. doi:10.1517/17425247.2012.642362. PMID 22251428. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  114. ^ Graber, JJ (1 June 2011). "Pharmacotherapy for primary CNS lymphoma: progress beyond methotrexate?". CNS drugs. 25 (6): 447–57. doi:10.2165/11589030-000000000-00000. PMID 21649446. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  115. ^ Dogra, S (August 2013). "Systemic methotrexate therapy for psoriasis: past, present and future". Clinical and Experimental Dermatology. 38 (6): 573–588. doi:10.1111/ced.12062. PMID 23837932. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  116. ^ Curtin, NJ (May 2001). "Pemetrexed disodium, a novel antifolate with multiple targets". The Lancet Oncology. 2 (5): 298–306. doi:10.1016/S1470-2045(00)00325-9. PMID 11905785. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  117. ^ Gatzemeier, U (November 2004). "Pemetrexed in malignant pleural mesothelioma". Oncology (Williston Park, N.Y.). 18 (13 Suppl 8): 26–31. PMID 15655933.
  118. ^ Sobrero, A (November 2004). "Pemetrexed in gastric cancer". Oncology (Williston Park, N.Y.). 18 (13 Suppl 8): 51–5. PMID 15655938. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  119. ^ Hazarika, M (1 February 2005). "Pemetrexed in malignant pleural mesothelioma" (PDF). Clinical cancer research: an official journal of the American Association for Cancer Research. 11 (3): 982–92. PMID 15709163. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  120. ^ Puto, K (April 2005). "Pemetrexed therapy for malignant pleural mesothelioma". The Annals of pharmacotherapy. 39 (4): 678–83. doi:10.1345/aph.1E329. PMID 15755794. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  121. ^ Villela, LR (May 2006). "Pemetrexed, a novel antifolate therapeutic alternative for cancer chemotherapy". Pharmacotherapy. 26 (5): 641–54. doi:10.1592/phco.26.5.641. PMID 16637794. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  122. ^ Goeminne, H (May 2006). "Pemetrexed in thoracic cancer". Expert opinion on pharmacotherapy. 7 (7): 917–28. doi:10.1517/14656566.7.7.917. PMID 16634714. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  123. ^ Monnerat, C (May 2006). "Review of the pemetrexed and gemcitabine combination in patients with advanced-stage non-small cell lung cancer" (PDF). Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 17 Suppl 5: v86-90. doi:10.1093/annonc/mdj958. PMID 16807472. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  124. ^ Longo-Sorbello, GS (February 2007). "Role of pemetrexed in non-small cell lung cancer". Cancer Investigation. 25 (1): 59–66. doi:10.1080/07357900601130748. PMID 17364559. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  125. ^ Chattopadhyay, S (February 2007). "Pemetrexed: Biochemical and cellular pharmacology, mechanisms, and clinical applications". Molecular Cancer Therapeutics. 6 (2): 404–417. doi:10.1158/1535-7163.MCT-06-0343. PMID 17308042. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  126. ^ Meriggi, F (December 2007). Chemotherapy. 54 (1): 1–8. doi:10.1159/000112311. PMID 18063861. {{cite journal}}: Missing or empty |title= (help); Unknown parameter |coauthors= ignored (|author= suggested) (help)
  127. ^ Kulkarni, PM (July 2008). "Efficacy and safety of pemetrexed in elderly cancer patients: results of an integrated analysis". Critical reviews in oncology/hematology. 67 (1): 64–70. doi:10.1016/j.critrevonc.2008.01.011. PMID 18358737. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  128. ^ Kulkarni, PM (July 2008). "Efficacy and safety of pemetrexed in elderly cancer patients: results of an integrated analysis". Critical reviews in oncology/hematology. 67 (1): 64–70. doi:10.1016/j.critrevonc.2008.01.011. PMID 18358737. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  129. ^ Felip, E (June 2008). "Pemetrexed as second-line therapy for advanced non-small-cell lung cancer (NSCLC)" (PDF). Therapeutics and clinical risk management. 4 (3): 579–85. doi:10.2147/TCRM.S2248. PMC 2500250. PMID 18827853. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  130. ^ Hsu, JY (September 2008). "Pemetrexed disodium for the treatment of NSCLC: an update". Drugs of today (Barcelona, Spain: 1998). 44 (9): 669–78. doi:10.1358/dot.2008.44.9.1250412. PMID 19137122. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  131. ^ Peake, MD (July 2009). "Pemetrexed in the treatment of malignant pleural mesothelioma". Therapy. 6 (4): 569–575. doi:10.2217/thy.09.30.
  132. ^ Manegold, C (September 2009). "Pemetrexed for the treatment of non-small-cell lung cancer". Expert review of anticancer therapy. 9 (9): 1195–209. doi:10.1586/ERA.09.97. PMID 19761423. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  133. ^ Tomao, F (December 2009). "Emerging role of pemetrexed in ovarian cancer". Expert review of anticancer therapy. 9 (12): 1727–35. doi:10.1586/ERA.09.141. PMID 19954283. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  134. ^ Fleeman, N (May 2010). "Pemetrexed for the first-line treatment of locally advanced or metastatic non-small cell lung cancer". Health technology assessment (Winchester, England). 14 Suppl 1: 47–53. doi:10.3310/hta14Suppl1/07. PMID 20507803. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  135. ^ Fuld, AD (June 2010). "Pemetrexed in advanced non-small-cell lung cancer". Expert opinion on pharmacotherapy. 11 (8): 1387–402. doi:10.1517/14656566.2010.482560. PMID 20446853. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  136. ^ Somer, R (April 2010). "Maintenance therapy for metastatic non-small-cell lung cancer – the role of pemetrexed" (PDF). Lung Cancer: Targets and Therapy. 1: 1–7. doi:10.2147/LCTT.S7105.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  137. ^ Greenhalgh, J (October 2010). "Pemetrexed for the maintenance treatment of locally advanced or metastatic non-small cell lung cancer". Health technology assessment (Winchester, England). 14 (Suppl. 2): 33–39. doi:10.3310/hta14suppl2/05. PMID 21047489. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  138. ^ Gridelli, C (March 2011). "Pemetrexed in advanced non-small cell lung cancer". Expert opinion on drug safety. 10 (2): 311–7. doi:10.1517/14740338.2011.553281. PMID 21261558. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  139. ^ Morotti, M (April 2012). "Pemetrexed disodium in ovarian cancer treatment". Expert opinion on investigational drugs. 21 (4): 437–49. doi:10.1517/13543784.2012.661714. PMID 22324304. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  140. ^ Argiris, A (June 2013). "Pemetrexed in head and neck cancer: a systematic review". Oral oncology. 49 (6): 492–501. doi:10.1016/j.oraloncology.2013.01.007. PMID 23466170. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  141. ^ Boons, CC (September 2013). "The value of pemetrexed for the treatment of malignant pleural mesothelioma: a comprehensive review". Anticancer research. 33 (9): 3553–61. PMID 24023280. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  142. ^ Gunasekara, NS (March 1998). "Raltitrexed. A review of its pharmacological properties and clinical efficacy in the management of advanced colorectal cancer". Drugs. 55 (3): 423–35. doi:10.2165/00003495-199855030-00012. PMID 9530547. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  143. ^ Clarke, SJ (December 2000). "Clinical and preclinical pharmacokinetics of raltitrexed". Clinical pharmacokinetics. 39 (6): 429–443. doi:10.1517/13543784.7.5.823. PMID 11192475. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  144. ^ Caponigro, F (July 2001). "Raltitrexed/5-fluorouracil-based combination chemotherapy regimens in anticancer therapy". Anti-cancer drugs. 12 (6): 489–97. doi:10.1097/00001813-200107000-00001. PMID 11459994. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  145. ^ Caponigro, F (July 2001). "Raltitrexed/5-fluorouracil-based combination chemotherapy regimens in anticancer therapy". Anti-cancer drugs. 12 (6): 489–97. PMID 11459994. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  146. ^ Cunningham, D (March 2002). "Efficacy, tolerability and management of raltitrexed (Tomudex™) monotherapy in patients with advanced colorectal cancer". European Journal of Cancer. 38 (4): 478–486. doi:10.1016/S0959-8049(01)00413-0. PMID 11872339. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  147. ^ Van Cutsem, E (1 April 2002). "Raltitrexed: current clinical status and future directions". Annals of Oncology. 13 (4): 513–522. doi:10.1093/annonc/mdf054. PMID 12056700. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  148. ^ Cao, S (April 2006). "Irinotecan, oxaliplatin and raltitrexed for the treatment of advanced colorectal cancer". Expert opinion on pharmacotherapy. 7 (6): 687–703. doi:10.1517/14656566.7.6.687. PMID 16556086. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  149. ^ Wilson, KS (November 2009). "Raltitrexed: optimism and reality". Expert opinion on drug metabolism & toxicology. 5 (11): 1447–1454. doi:10.1517/17425250903307455. PMID 19863453. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  150. ^ Mulaku, M (November 2013). "Evidence review of hydroxyurea for the prevention of sickle cell complications in low-income countries" (PDF). Archives of Disease in Childhood. 98 (11): 908–14. doi:10.1136/archdischild-2012-302387. PMC 3812872. PMID 23995076. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  151. ^ Navarra, P (February 1999). "Hydroxyurea: new insights on an old drug". Critical reviews in oncology/hematology. 29 (3): 249–55. doi:10.1016/S1040-8428(98)00032-8. PMID 10226728. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  152. ^ Romanelli, F (February 1999). "Hydroxyurea to inhibit human immunodeficiency virus-1 replication". Pharmacotherapy. 19 (2): 196–204. doi:10.1592/phco.19.3.196.30913. PMID 10030769. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  153. ^ Vichinsky, EP (July 1997). "Hydroxyurea in children: present and future". Seminars in Hematology. 34 (3 Suppl 3): 22–9. PMID 9317198.
  154. ^ Gwilt, PR (May 1998). "Pharmacokinetics and pharmacodynamics of hydroxyurea". Clinical pharmacokinetics. 34 (5): 347–358. doi:10.2165/00003088-199834050-00002. PMID 9592619. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  155. ^ Ravot, Elisabetta (1999). "New Uses for Old Drugs in HIV Infection". Drugs. 58 (6): 953–963. doi:10.2165/00003495-199958060-00001. PMID 10651384. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  156. ^ "Hydroxyurea" (PDF). 76. 2000. {{cite journal}}: Cite journal requires |journal= (help)
  157. ^ Banan, M (March 2013). "Hydroxyurea treatment in β-thalassemia patients: to respond or not to respond?". Annals of hematology. 92 (3): 289–99. doi:10.1007/s00277-012-1671-3. PMID 23318979.
  158. ^ Dingli, D (June 2006). "Hydroxyurea: The drug of choice for polycythemia vera and essential thrombocythemia". Current Hematologic Malignancy Reports. 1 (2): 69–74. doi:10.1007/s11899-006-0025-4. PMID 20425334. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  159. ^ Lisziewicz, J (2003). "Hydroxyurea in the treatment of HIV infection: clinical efficacy and safety concerns". Drug safety. 26 (9): 605–24. doi:10.2165/00002018-200326090-00002. PMID 12814330. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  160. ^ a b c d e f g h i Bhatia, S (2008). "Secondary Malignancies: Therapy-Related t-MDS/AML". Medscape Reference. WebMD. Retrieved 7 February 2014.
  161. ^ Shimada, M (May 2013). "Nedaplatin: a cisplatin derivative in cancer chemotherapy" (PDF). Cancer management and research. 5: 67–76. doi:10.2147/CMAR.S35785. PMC 3658440. PMID 23696716. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  162. ^ Ravandi, F (17 September 2012). "Gemtuzumab Ozogamicin: Time to Resurrect?" (PDF). Journal of Clinical Oncology. 30 (32): 3921–3923. doi:10.1200/JCO.2012.43.0132. PMID 22987091. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  163. ^ Lehnert, M (February 2009). "Update on the rational use of Y-ibritumomab tiuxetan in the treatment of follicular lymphoma" (PDF). OncoTargets and therapy. 2: 199–208. PMC 2886339. PMID 20616907. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  164. ^ Cheson, BD (2005). "The role of radioimmunotherapy with yttrium-90 ibritumomab tiuxetan in the treatment of non-Hodgkin lymphoma". BioDrugs: clinical immunotherapeutics, biopharmaceuticals and gene therapy. 19 (5): 309–22. doi:10.2165/00063030-200519050-00004. PMID 16207072.
  165. ^ Nabhan, C (March 2011). "The emerging role of ofatumumab in the treatment of chronic lymphocytic leukemia" (PDF). Clinical Medicine Insights. Oncology. 5: 45–53. doi:10.4137/CMO.S4087. PMC 3076040. PMID 21499555. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  166. ^ Osterborg, A (March 2010). "Ofatumumab, a human anti-CD20 monoclonal antibody". Expert Opinion on Biological Therapy. 10 (3): 439–49. doi:10.1517/14712590903586239. PMID 20109133.
  167. ^ Sanford, M (May 2010). "Ofatumumab". Drugs. 70 (8): 1013–1019. doi:10.2165/11203850-000000000-00000. PMID 20481657. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  168. ^ Davies, A J (28 May 2007). "Radioimmunotherapy for B-cell lymphoma: Y90 ibritumomab tiuxetan and I131 tositumomab". Oncogene. 26 (25): 3614–3628. doi:10.1038/sj.onc.1210378. PMID 17530015.
  169. ^ "Bexxar (tositumomab) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  170. ^ "Gilotrif (afatinib)". Medscape Reference. WebMD. Retrieved 9 February 2014.
  171. ^ "Zaltrap (ziv-aflibercept) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  172. ^ Escudier, B (2011). "Axitinib for the management of metastatic renal cell carcinoma" (PDF). Drugs in R&D. 11 (2): 113–26. doi:10.2165/11591240-000000000-00000. PMC 21679004. PMID 21679004. {{cite journal}}: Check |pmc= value (help); Unknown parameter |coauthors= ignored (|author= suggested) (help)
  173. ^ "Bosulif (bosutinib) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  174. ^ Koschmieder, S (March 2013). "Profile of bosutinib and its clinical potential in the treatment of chronic myeloid leukemia" (PDF). OncoTargets and Therapy. 6: 99–106. doi:10.2147/OTT.S19901. PMC 3594007. PMID 23493838. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  175. ^ "Xalkori (crizotinib) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  176. ^ Forde, PM (June 2012). "Crizotinib in the treatment of non-small-cell lung cancer". Expert opinion on pharmacotherapy. 13 (8): 1195–201. doi:10.1517/14656566.2012.688029. PMID 22594847. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  177. ^ Frampton, JE (28 November 2013). "Crizotinib: A Review of Its Use in the Treatment of Anaplastic Lymphoma Kinase-Positive, Advanced Non-Small Cell Lung Cancer". Drugs. 73 (18): 2031–2051. doi:10.1007/s40265-013-0142-z. PMID 24288180.
  178. ^ Rothschild, SI (September 2013). "Crizotinib in the Treatment of Non–Small-Cell Lung Cancer". Clinical Lung Cancer. 14 (5): 473–480. doi:10.1016/j.cllc.2013.04.006. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  179. ^ Roskoski, R (September 2013). "The preclinical profile of crizotinib for the treatment of non-small-cell lung cancer and other neoplastic disorders". Expert Opinion on Drug Discovery. 8 (9): 1165–1179. doi:10.1517/17460441.2013.813015. PMID 23805942.
  180. ^ "Iclusig (ponatinib) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  181. ^ Price, KE (August 2013). "Potential of ponatinib to treat chronic myeloid leukemia and acute lymphoblastic leukemia" (PDF). OncoTargets and therapy. 6: 1111–8. doi:10.2147/OTT.S36980. PMC 3754816. PMID 23986642. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)
  182. ^ "Stivarga (regorafenib) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  183. ^ Strumberg, D (June 2012). "Regorafenib for cancer". Expert opinion on investigational drugs. 21 (6): 879–889. doi:10.1517/13543784.2012.684752. PMID 22577890. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  184. ^ Aprile, G (23 February 2013). "Regorafenib for Gastrointestinal Malignancies". BioDrugs. 27 (3): 213–224. doi:10.1007/s40259-013-0014-9. PMID 23435872. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  185. ^ Shahda, S (May 2013). "Regorafenib: from bench to bedside in colorectal cancer". Expert Review of Clinical Pharmacology. 6 (3): 243–248. doi:10.1586/ecp.13.11. PMID 23656338. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  186. ^ "Jakafi (ruxolitinib) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 10 February 2014.
  187. ^ "JAKAVI® Ruxolitinib" (PDF). TGA eBusiness Services. Novartis Pharmaceuticals Australia Pty Limited. 21 January 2014.
  188. ^ Kantarjian, HM (December 2013). "Ruxolitinib for Myelofibrosis–An Update of Its Clinical Effects" (PDF). Clinical Lymphoma Myeloma and Leukemia. 13 (6): 638–645. doi:10.1016/j.clml.2013.09.006. PMID 24238036. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  189. ^ Vaddi, K (November 2012). "Ruxolitinib, an oral JAK1 and JAK2 inhibitor, in myelofibrosis". Expert Opinion on Pharmacotherapy. 13 (16): 2397–2407. doi:10.1517/14656566.2012.732998. PMID 23051187. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  190. ^ "Vandetanib dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  191. ^ "Targretin (bexarotene) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  192. ^ Matthay, KK (3 December 2012). "Targeted Isotretinoin in Neuroblastoma: Kinetics, Genetics, or Absorption". Clinical Cancer Research. 19 (2): 311–313. doi:10.1158/1078-0432.CCR-12-3313. PMID 23209029.
  193. ^ "Amnesteem, Claravis (isotretinoin) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  194. ^ "Isotretinoin 20mg capsules - Summary of Product Characteristics (SPC)". electronic Medicines Compendium. Alliance Pharmaceuticals. 12 April 2013. Retrieved 9 February 2014.
  195. ^ Miwako, I (August 2007). "Tamibarotene". Drugs of today (Barcelona, Spain: 1998). 43 (8): 563–8. doi:10.1358/dot.2007.43.8.1072615. PMID 17925887. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  196. ^ Fukasawa, H (2012). "Tamibarotene: a candidate retinoid drug for Alzheimer's disease". Biological & pharmaceutical bulletin. 35 (8): 1206–12. doi:10.1248/bpb.b12-00314. PMID 22863914. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  197. ^ Takeuchi, M (March 2006). "[Clinical experience with a new synthetic retinoid, tamibarotene (Am-80) for relapsed or refractory acute promyelocytic leukemia]". Gan to kagaku ryoho. Cancer & chemotherapy (in Japanese). 33 (3): 397–401. PMID 16531727.
  198. ^ "Istodax (romidepsin) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 10 February 2014.
  199. ^ Bellos, F (November 2008). "Valproic acid and all-trans retinoic acid: meta-analysis of a palliative treatment regimen in AML and MDS patients". Onkologie. 31 (11): 629–33. doi:10.1159/000160599. PMID 19145098. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  200. ^ Blaheta, RA (August 2002). "Valproate and valproate-analogues: potent tools to fight against cancer". Current medicinal chemistry. 9 (15): 1417–33. doi:10.2174/0929867023369763. PMID 12173980. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  201. ^ Blaheta, RA (September 2002). "Anti-tumor mechanisms of valproate: a novel role for an old drug". Medicinal research reviews. 22 (5): 492–511. doi:10.1002/med.10017. PMID 12210556. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  202. ^ Duenas-Gonzalez, A (May 2008). "Valproic acid as epigenetic cancer drug: preclinical, clinical and transcriptional effects on solid tumors". Cancer treatment reviews. 34 (3): 206–22. doi:10.1016/j.ctrv.2007.11.003. PMID 18226465. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  203. ^ "Zolinza (vorinostat) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 10 February 2014.
  204. ^ "Ontak (denileukin) dosing, indications, interactions, adverse effects, and more". Medscape Reference. WebMD. Retrieved 9 February 2014.
  205. ^ Lansigan, F (February 2010). "Role of denileukin diftitox in the treatment of persistent or recurrent cutaneous T-cell lymphoma" (PDF). Cancer Management and Research. 2: 53–59. doi:10.2147/CMAR.S5009. PMC 3004568. PMID 21188096.{{cite journal}}: CS1 maint: unflagged free DOI (link)
  206. ^ Manoukian, G (November 2009). "Denileukin diftitox: a novel immunotoxin". Expert Opinion on Biological Therapy. 9 (11): 1445–1451. doi:10.1517/14712590903348135. PMID 19817678. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)
  207. ^ Hymes, KB (December 2008). "Denileukin diftitox for the treatment of cutaneous T-cell lymphoma" (PDF). Biologics: Targets & Therapy. 2 (4): 717–724. doi:10.2147/BTT.S3084. PMC 2727893. PMID 19707452. {{cite journal}}: Unknown parameter |coauthors= ignored (|author= suggested) (help)CS1 maint: unflagged free DOI (link)