Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
N Z Med J ; 134(1533): 33-45, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33927422

ABSTRACT

BACKGROUND: Chemotherapy-induced nausea and vomiting (CINV) are two of the most frequently experienced and distressing side effects of cancer treatment. Recent updates by ESMO/MASCC and ASCO on guidelines for prevention of CINV have recommended the addition of a neurokinin-1 receptor antagonist to antiemetic regimens for patients receiving carboplatin-based chemotherapy area under the curve (AUC) ≥4 mg/mL per minute, and an addition of olanzapine for those receiving combination anthracycline/cyclophosphamide chemotherapy. AIMS: To assess current use of prophylactic antiemetics and rates of CINV in patients under the care of MidCentral Regional Cancer Treatment Service (MRCTS) receiving carboplatin AUC≥4 or combination anthracycline/cyclophosphamide. METHODS: Data was prospectively collected on patients under the care of MRCTS receiving carboplatin AUC≥4 or combination anthracycline/cyclophosphamide chemotherapy, including breast cancer patients receiving 5-fluorouracil, epirubicin and cyclophosphamide (FEC) and lymphoma patients receiving rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP). Questionnaires were given to eligible patients to be completed daily from day two to day six of first cycle of chemotherapy only. Data on each patient's gender, age, types of chemotherapies, types of malignancies, presence of nausea or vomiting, number of dry retching or vomiting episodes and anti-emetics were recorded. RESULTS: From 15 September 2018 to 10 August 2019, a total of 44 patients receiving carboplatin-based chemotherapy AUC≥4 and 30 patients receiving combination anthracycline/cyclophosphamide were included. Twenty-two patients (50%) had either emesis or significant nausea in the overall and delayed phase when treated with carboplatin AUC≥4, and only three (7%) in the acute phase. Fourteen patients (56%) had either emesis or significant nausea in the overall phase when treated with FEC chemotherapy, mostly in the acute phase (13 patients) rather than in the delayed phase (9 patients). CONCLUSION: The rates of CINV are high with the existing antiemetic regimens used at MidCentral Regional Cancer Treatment Service. Therefore, in accordance with international guidelines, we will add a neurokinin-1 antagonist to the antiemetic regimens for patients receiving carboplatin-based chemotherapy AUC≥4, and olanzapine for those receiving combination anthracycline/cyclophosphamide chemotherapy, in an attempt to improve the rates of CINV in these groups. Repeating this audit post-implementation of above recommendations will be important to assess for any improvement.


Subject(s)
Anthracyclines/adverse effects , Antiemetics/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carboplatin/adverse effects , Cyclophosphamide/adverse effects , Nausea/prevention & control , Vomiting/prevention & control , Adult , Aged , Aged, 80 and over , Anthracyclines/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Area Under Curve , Carboplatin/pharmacokinetics , Cyclophosphamide/pharmacokinetics , Female , Humans , Male , Medical Audit , Middle Aged , Prospective Studies
2.
Cancer Chemother Pharmacol ; 88(2): 203-209, 2021 08.
Article in English | MEDLINE | ID: mdl-33907881

ABSTRACT

PURPOSE: As critical parameter after extravasation of cytotoxic vesicants, anthracyclines were determined in removed tissue from patients requiring surgical intervention due to tissue necrosis. We monitored their distribution within the affected lesion to establish a possible dose-toxicity relation. METHODS: From six patients scheduled for surgery, removed tissue flaps were systematically analysed by HPLC (epirubicin: 5 subjects; doxorubicin: 1 subject). RESULTS: After extravasation, tissue concentrations were highly variable with an individual anthracycline distribution pattern ranging from a few nanograms up to 17 µg per 100 mg tissue, which indicated a substantial difference in tissue sensitivity among patients. The resection borders coincided with the extension of the erythema and guided the surgical intervention after demarcation of the lesion, which occurred usually 2 or 3 weeks after extravasation. At that time, drug was hardly detected at the resection borders. Wound drains were negative for the extravasated drugs while showing a time profile of vascular growth factors and inflammatory cytokines, which was highly similar to routine surgery. In all six patients, surgical debridement with immediate wound closure led to healing within approximately 2 weeks, when therapy was resumed in all patients with reasonable time delay. CONCLUSION: Surgical intervention after demarcation of the extravasation lesion allows for almost uninterrupted continuation of treatment independent of the amount of extravasated anthracycline. As even minor amounts of the vesicants may trigger tissue necrosis, preventive measures merit the highest priority.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacokinetics , Epirubicin/adverse effects , Epirubicin/pharmacokinetics , Tissue Distribution/physiology , Aged , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Anthracyclines/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cytokines/metabolism , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Epirubicin/therapeutic use , Female , Humans , Inflammation/metabolism , Male , Middle Aged , Necrosis/chemically induced , Necrosis/metabolism , Skin Diseases/drug therapy , Skin Diseases/metabolism , Surgical Flaps/pathology , Wound Healing/drug effects
3.
Invest New Drugs ; 39(2): 530-536, 2021 04.
Article in English | MEDLINE | ID: mdl-33159674

ABSTRACT

Background Amrubicin (AMR) is a completely synthetic 9-aminoanthracycline and clinically active against non-small cell lung cancer (NSCLC). We conducted a phase I study of AMR and erlotinib (ERL) combination therapy in previously treated patients with advanced NSCLC and have already reported the safety and effectiveness. Methods We conducted a multi-center, single-arm phase II trial to evaluate the efficacy of AMR and ERL combination therapy in patients with previously treated, advanced NSCLC harboring wild-type EGFR, PS 0-1 and < 75 years of age. Patients were treated at 3-week intervals with AMR plus ERL. The primary endpoint was the PFS, and the secondary endpoints were the response rate (RR), disease control rate (DCR), overall survival (OS) and toxicity. The trough ERL concentration (Ctrough) was measured as an exploratory study to analyze the relationship between the efficacy/safety and pharmacokinetics. Results From June 2013 to July 2016, 25 patients were enrolled in this trial. The PFS according to the central test was 3.6 months (95% confidence interval 2.1-5.1). The RR and DCR were 24.0% and 64.0%, respectively. We had no treatment-related deaths in this study. Conclusions The PFS of AMR and ERL combination therapy was superior to that of AMR monotherapy in the historical setting, but the primary endpoint was not met in this trial. In our study, the pharmacokinetic analysis showed that the Ctrough of ERL was elevated with combination therapy. This combination therapy might be a viable treatment for previously treated NSCLC patients without a driver oncogene mutation. Clinical trial information UMIN 000010582.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Erlotinib Hydrochloride/therapeutic use , Lung Neoplasms/drug therapy , Adult , Aged , Anthracyclines/administration & dosage , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/adverse effects , Erlotinib Hydrochloride/pharmacokinetics , Humans , Lung Neoplasms/genetics , Male , Middle Aged , Progression-Free Survival
4.
Biochem Pharmacol ; 177: 113959, 2020 07.
Article in English | MEDLINE | ID: mdl-32272110

ABSTRACT

Breast cancer is the most prevalent type of tumor and the second leading cause of death due to cancer among women. Although screening methods, diagnosis and therapeutic options have improved in the last decade, chemoresistance remains an important challenge. There is evidence relating breast cancer resistance with signaling pathways involving hormone and growth receptors, survival, apoptosis and the activation of efflux pumps. However, the resistance mechanisms linked to drug uptake are poorly understood, despite it often being observed that the drug content is lower in resistant cancer cells and that the entry of the drug into these cells is a limiting process for the subsequent therapeutic effect.In this review, we provide an overview of drug uptake-based resistance mechanisms developed by cancer cells in the four main types of chemotherapy used in breast cancer: anthracyclines, taxanes, oxazaphosphorines and platinum-based drugs. The contribution of tumor microenvironment to reduced drug-uptake and multidrug resistance is also analyzed. As a developing field, nanomedicine-based approaches provide promising opportunities to improve drug specific targeting, cell interaction and uptake into cancer cells. The endocytic-mediated pathways attributed to the different types of nanoformulations as well as the contribution of nanotherapeutics to overcoming chemoresistance affecting drug uptake in breast cancer will be described. New approaches focusing on drug uptake mechanisms could improve breast cancer chemotherapy, obtaining better dose-response outcomes and reducing toxic side effects.


Subject(s)
Anthracyclines/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Oxazines/therapeutic use , Platinum Compounds/therapeutic use , Taxoids/therapeutic use , Anthracyclines/pharmacokinetics , Breast Neoplasms/metabolism , Female , Humans , Nanomedicine/methods , Nanomedicine/trends , Oxazines/pharmacokinetics , Platinum Compounds/pharmacokinetics , Taxoids/pharmacokinetics , Tumor Microenvironment/drug effects
5.
Cancer Sci ; 110(11): 3573-3583, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31505087

ABSTRACT

This study determined individual optimal amrubicin doses for Japanese patients with lung cancer after platinum-based treatment. We carried out population pharmacokinetic and pharmacodynamic modeling incorporating gene polymorphisms of metabolizing enzymes and transporters. Fifty patients with lung cancer, who were given 35-40 mg/m2 amrubicin on days 1-3 every 3-4 weeks, were enrolled. Mechanism-based modeling described relationships between the pharmacokinetics of amrubicin and absolute neutrophil counts. A population pharmacokinetic and pharmacodynamic model was developed for amrubicin and amrubicinol (active metabolite), connected by a delay compartment. The final model incorporated body surface area as a covariate of amrubicin and amrubicinol clearance and distribution volume. SLC28A3 single nucleotide polymorphism (rs7853758) was also incorporated as a constant covariate of the delay compartment of amrubicinol. Performance status was considered a covariate of pharmacokinetic (amrubicinol clearance) and pharmacodynamic (mean maturation time) parameters. Twenty-nine patients with grade 4 neutropenia showed higher amrubicinol area under the plasma concentration-time curve from 0 to 72 hours (AUC0-72 , P = .01) and shorter overall survival periods than other patients did (P = .01). Using the final population pharmacokinetic and pharmacodynamic model, median optimal dose to prevent grade 4 neutropenia aggravation was estimated at 22 (range, 8-40) mg/m2 for these 29 patients. We clarified correlations between area under the plasma concentration-time curve from 0 to 72 hours of amrubicinol and severity of neutropenia and survival of patients given amrubicin after platinum chemotherapy. This analysis revealed important amrubicin pharmacokinetic-pharmacodynamic covariates and provided useful information to predict patients who would require prophylactic granulocyte colony stimulating factor.


Subject(s)
Anthracyclines/administration & dosage , Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neutropenia/prevention & control , Adult , Aged , Aged, 80 and over , Anthracyclines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Asian People , Body Surface Area , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Drug Administration Schedule , Female , Humans , Japan , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Membrane Transport Proteins/genetics , Middle Aged , Neutropenia/chemically induced , Neutropenia/metabolism , Polymorphism, Single Nucleotide , Prospective Studies , Severity of Illness Index
6.
Nanomedicine (Lond) ; 14(13): 1663-1680, 2019 07.
Article in English | MEDLINE | ID: mdl-31167626

ABSTRACT

Aim: This study aimed to develop anthracycline-loaded bacterial magnetosomes (BMs) with enhanced anticancer efficiency and elucidate their endocytosis mechanism. Methods: Drug-loaded BMs (DBMs) were successfully prepared and characterized. DBMs endocytosis was investigated within HepG2 cells. The anticancer effect of DBMs was studied both in vitro and in vivo. Results: Doxorubicin-BMs and daunorubicin-BMs showed enhanced growth inhibitory effect in vitro and in vivo with no notable toxicity to normal tissues. The DBMs were internalized into cells through caveolae-mediated endocytosis and macropinocytosis. The loaded drugs were released from DBMs in cytoplasm and entered the nucleus to exert their activity. Conclusion: Our findings offer promising candidates for improved cancer therapy with a clear mechanism of DBMs endocytosis and working principle.


Subject(s)
Anthracyclines/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Drug Carriers/chemistry , Liver Neoplasms/drug therapy , Magnetosomes/chemistry , Anthracyclines/pharmacokinetics , Anthracyclines/pharmacology , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Endocytosis , Hep G2 Cells , Humans , Liver Neoplasms/metabolism
7.
Anticancer Drugs ; 29(9): 821-826, 2018 10.
Article in English | MEDLINE | ID: mdl-30036190

ABSTRACT

The extravasation of chemotherapeutic agents is a challenge for oncologic care teams. The management of nonliposomal (conventional) anthracyclines is well established in clinical practice guidelines, including general measures and specific antidotes, such as dexrazoxane. However, there is little scientific evidence on the management of liposomal and pegylated liposomal anthracyclines. The aim of this paper was to review the scientific literature on the extravasation of liposomal and pegylated liposomal anthracyclines and determine the clinical impact of this type of extravasation, focusing on dexrazoxane. The literature was searched using two databases: PubMed and Embase. Three searches were conducted, using liposomal anthracycline extravasation, pegylated liposomal anthracycline extravasation, and liposomal doxorubicin extravasation as keywords, respectively. Seven articles fulfilled the study eligibility criteria and included seventeen cases in humans. Extravasation occurred with three drugs: liposomal doxorubicin in nine (53%) patients, liposomal daunorubicin in four (23.5%) patients, and pegylated liposomal doxorubicin in four (23.5%) patients. General measures for extravasations were applied in all patients, but only three patients received dexrazoxane. All cases were completely resolved at 2-3 months, except for one patient, in whom dexrazoxane was not used. In animals, dexrazoxane decreased both the frequency of wounds produced by pegylated liposomal doxorubicin and their extent. The pharmacokinetic profiles of liposomal and pegylated liposomal anthracyclines differ from those of conventional anthracyclines, modifying their effectiveness and safety. General measures may be inadequate to heal areas affected by extravasation, which may require the administration of dexrazoxane. However, each case should be evaluated individually for the administration of dexrazoxane in off-label use until scientific evidence is available on its effectiveness and safety as an antidote for these formulations of anthracyclines.


Subject(s)
Anthracyclines/administration & dosage , Dexrazoxane/administration & dosage , Extravasation of Diagnostic and Therapeutic Materials/drug therapy , Animals , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacokinetics , Daunorubicin/administration & dosage , Daunorubicin/adverse effects , Daunorubicin/pharmacokinetics , Dexrazoxane/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Extravasation of Diagnostic and Therapeutic Materials/etiology , Humans , Liposomes , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Polyethylene Glycols/pharmacokinetics
8.
Curr Treat Options Oncol ; 19(6): 30, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29752560

ABSTRACT

OPINION STATEMENT: Anthracycline-based regimens have been an important treatment component for patients with breast cancer. As demonstrated in the last Early Breast Cancer Trialists' Collaborative Group (EBCTCG) meta-analysis, anthracycline-based regimens decrease breast cancer mortality by 20-30%. Anthracycline toxicities include the rare-but potential morbid-cardiotoxicity or leukemogenic effect, and the almost universal-but very distressing-alopecia. Due to potential toxicities, and large number of patients being exposed, several worldwide trials have re-examined the role of anthracycline-based regimens in the management of breast cancer. Current literature supports that anthracyclines are not required for all patients with breast cancer and should be avoided in those with high cardiac risk. Recent results from the ABC trials suggest that anthracyclines should not be spared for patients with triple negative breast cancer (regardless of axillary node involvement) or HER2-/ER+ with significant node involvement. Based on current literature, for HER2-negative patients with low-risk breast cancer, anthracyclines could be spared with regimens such as cyclophosphamide, methotrexate, and fluorouracil (CMF) or docetaxel and cyclophosphamide (TC). Patients with intermediate or high-risk breast cancer should be considered for anthracycline-based regimens based on other factors such as age, comorbidities, tumor grade, lymphovascular invasion, and genomic profiling. Patients with HER2-positive breast cancer with low risk could be treated with paclitaxel and trastuzumab. For the remaining patients with HER2 overexpression, while docetaxel, carboplatin, and trastuzumab (TCH) has demonstrated to improve disease-free survival (DFS), anthracycline-containing regimens should be discussed, especially for those with very high-risk breast cancer. Although several biomarkers, such as topoisomerase II (TOP2A) and chromosome 17 centromeric duplication (Ch17CEP) have been proposed to predict benefit from anthracycline regimens, further research is required to delineate their proper utility in the clinical setting.


Subject(s)
Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Anthracyclines/administration & dosage , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Breast Neoplasms/etiology , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Clinical Trials as Topic , Disease Management , Female , Humans , Neoplasm Staging , Prognosis , Treatment Outcome
9.
J Pharmacol Exp Ther ; 364(2): 323-331, 2018 02.
Article in English | MEDLINE | ID: mdl-29222131

ABSTRACT

Cumulative doses of doxorubicin and other antitumor anthracyclines may cause heart failure (HF). Cardiotoxicity is determined by cardiac exposure to anthracyclines and to more toxic secondary alcohol metabolites that are formed inside cardiomyocytes or diffuse from the bloodstream. Concerns exist that HF might be caused by cumulative anthracycline doses that were thought to be safe. Patients with gain-of-function polymorphism of carbonyl reductase 3 (CBR3), which converts anthracyclines to secondary alcohol metabolites, would be at a higher risk of HF. Recently, a pharmacokinetic model was developed that simulated clinical exposure of human myocardium to anthracyclines and incorporated simulations of CBR3 polymorphism. It was shown that HF risk could occur after lower doxorubicin doses than previously reported, particularly for patients with CBR3 polymorphism. In this study, we show that also daunorubicin and idarubicin, but not epirubicin, might cause HF after reportedly safe cumulative doses. CBR3 polymorphism increased HF risk from daunorubicin and idarubicin to a greater extent as compared with doxorubicin. This was caused by daunorubicin and idarubicin forming higher levels of toxic metabolites in human myocardium; moreover, daunorubicin and idarubicin metabolites diffused from plasma and accumulated in cardiac tissue, whereas doxorubicin metabolite did not. CBR3 polymorphism did not aggravate HF risk from epirubicin, which was caused by the very low levels of formation of its toxic metabolite. These results support concerns about HF risk from low-dose anthracycline, characterize the analog specificity of HF risk, and illuminate the role of secondary alcohol metabolites.


Subject(s)
Alcohols/metabolism , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Heart Failure/chemically induced , Heart Failure/metabolism , Heart/drug effects , Myocardium/metabolism , Adult , Aged , Aged, 80 and over , Alcohol Oxidoreductases/metabolism , Conotoxins , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged
10.
J Transl Med ; 15(1): 235, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29141648

ABSTRACT

BACKGROUND: Gene polymorphisms at microRNA-binding sites (poly-miRTS) may affect gene transcription and expression through miRNA regulation, which is associated with cancer susceptibility, sensitivity to chemotherapy and prognosis. This study investigated the association between poly-miRTS of Ara-C/anthracycline metabolic pathways genes and the outcome of acute myeloid leukemia (AML) in Chinese patients after Ara-C-based chemotherapy. METHODS: A total of 17 poly-miRTS were selected from the SNPinfo Web Server and genotyped in 206 Chinese Han non-FAB-M3 AML patients using the SEQUENOM Mass-ARRAY system. RESULTS: Among these 17 poly-miRTS, five Ara-C metabolic gene single nucleotide polymorphisms (SNPs, NT5C2 rs10786736 and rs8139, SLC29A1 rs3734703, DCTD rs7278, and RRM1 rs1042919) were identified to significantly associate with complete AML remission and/or overall and relapse-free survival (OS and RFS, respectively), and four anthracycline-metabolic gene SNPs (ABCC1 rs3743527, rs212091, and rs212090 and CBR1 rs9024) were significantly associated with chemotherapy-related toxicities. Moreover, SLC29A1 rs3734703 was shown to associate with both chemotherapy response and survival (adjusted OR 2.561 in the overdominant model; adjusted HR 2.876 for OS and 2.357 for RFS in the dominant model). CONCLUSIONS: The data from the current study demonstrated that the poly-miRTS of Ara-C/anthracyclines metabolic genes predicted the sensitivity and side effects of AML to Ara-C-based chemotherapy and patient survival. Further study will confirm them as biomarkers for AML patients after Ara-C-based chemotherapy.


Subject(s)
Anthracyclines/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cytarabine/metabolism , Inactivation, Metabolic/genetics , Leukemia, Myeloid, Acute , MicroRNAs , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Anthracyclines/administration & dosage , Binding Sites/genetics , Biomarkers, Tumor/genetics , Cytarabine/administration & dosage , Female , Follow-Up Studies , Genetic Predisposition to Disease , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Male , Metabolic Networks and Pathways/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Prognosis , Remission Induction , Survival Analysis , Treatment Outcome , Young Adult
11.
Nutr Cancer ; 69(4): 555-563, 2017.
Article in English | MEDLINE | ID: mdl-28353359

ABSTRACT

The etiology of undernourishment in cancer patients is multifactorial: tumor-related mechanisms (such as obstruction, metabolic abnormalities, and functionality changes) in addition to the influence of anticancer therapies, which can induce or worsen undernutrition. The evident role of undernutrition in cancer treatment outcomes suggests the need of considering nutritional status when evaluating anticancer drugs. In order to merge the available data and offer researchers and clinicians a global view of this phenomenon, the present manuscript reviews on a drug-by-drug basis the undernutrition-related pharmacokinetic and pharmacodynamic aspects of anticancer treatments. This review notes interesting trends in the relationship between undernourishment and pharmacokinetics across studies, and indicates that dosing modifications of these drugs may be necessary to optimize chemotherapeutic treatments. Furthermore, this review has compiled evidence regarding undernourishment's capacity of enhancing treatment-related myelosuppression, cardiotoxicity, ototoxicity, neurotoxicity, and malignancies.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Malnutrition/physiopathology , Anthracyclines/pharmacokinetics , Etoposide/pharmacokinetics , Fluorouracil/pharmacokinetics , Humans , Methotrexate/pharmacokinetics , Vinca Alkaloids/pharmacokinetics
12.
Invest New Drugs ; 35(3): 375-385, 2017 06.
Article in English | MEDLINE | ID: mdl-28283780

ABSTRACT

Anthracycline antibiotics (ANT), such as doxorubicin or daunorubicin, are a class of anticancer drugs that are widely used in oncology. Although highly effective in cancer therapy, their usefulness is greatly limited by their cardiotoxicity. Possible mechanisms of ANT cardiotoxicity include their conversion to secondary alcohol metabolites (i.e. doxorubicinol, daunorubicinol) catalyzed by carbonyl reductases (CBR) and aldo-keto reductases (AKR). These metabolites are suspected to be more cardiotoxic than their parent compounds. Moreover, overexpression of ANT-reducing enzymes (CBR and AKR) are found in many ANT-resistant cancers. The secondary metabolites show decreased cytotoxic properties and are more susceptible to ABC-mediated efflux than their parent compounds; thus, metabolite formation is considered one of the mechanisms of cancer resistance. Inhibitors of CBR and AKR were found to reduce the cardiotoxicity of ANT and the resistance of cancer cells, and therefore are being investigated as prospective cardioprotective and chemosensitizing drug candidates. In this review, the significance of a two-electron reduction of ANT, including daunorubicin, epirubicin, idarubicin, valrubicin, amrubicin, aclarubicin, and especially doxorubicin, is described with respect to toxicity and efficacy of therapy. Additionally, CBR and AKR inhibitors, including monoHER, curcumin, (-)-epigallocatechin gallate, resveratrol, berberine or pixantrone, and their modulating effect on the activity of ANT is characterized and discussed as potential mechanism of action for novel therapeutics in cancer treatment.


Subject(s)
Aldo-Keto Reductases/antagonists & inhibitors , Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Carbonyl Reductase (NADPH)/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Cardiotoxicity/prevention & control , Animals , Anthracyclines/pharmacokinetics , Antibiotics, Antineoplastic/pharmacokinetics , Cardiotoxicity/etiology , Drug Resistance, Neoplasm , Humans
13.
Pharmacogenomics ; 17(9): 1075-87, 2016 06.
Article in English | MEDLINE | ID: mdl-27266419

ABSTRACT

Anthracyclines constitute a fundamental part of the chemotherapy regimens utilized to treat a number of different malignancies both in pediatric and adult patients. These drugs are one of the most efficacious anticancer agents ever invented. On the other hand, anthracyclines are cardiotoxic. Childhood cancer survivors treated with anthracyclines often undergo cardiac complications which are influenced by genetic variations of the patients. The scientific literature comprises numerous investigations in the subject of the pharmacogenetics of anthracyclines. In this review, we provide a comprehensive overview of this research topic. Genetic variants are proposed targets in the personalized treatment in order to individualize dosing and therefore reduce side effects.


Subject(s)
Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Pharmacogenetics , Animals , Anthracyclines/pharmacokinetics , Antibiotics, Antineoplastic/pharmacokinetics , Humans
14.
Expert Opin Pharmacother ; 17(11): 1569-77, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27328277

ABSTRACT

INTRODUCTION: Trabectedin, a marine-derived DNA-binding antineoplastic agent, has been registered by the EMA and recently also by the FDA for the treatment of patients with advanced soft-tissue sarcoma (STS), a rare and heterogeneous disease. AREAS COVERED: The antitumor activity of trabectedin is related both to direct effects on cancer cells, such as growth inhibition, cell death and differentiation, and indirect effects related to its anti-inflammatory and anti-angiogenic properties. Furthermore, trabectedin is the first compound that targets an oncogenic transcription factor with high selectivity in mixoid liposarcomas. This peculiar mechanism of action is the basis of its clinical development. The clinical pharmacology of trabectedin, the subsequent phase I, II and III trials are summarized and put into perspectives in this review. EXPERT OPINION: Trabectedin is a relevant pleiotropic antitumoral agent within the complex scenario of the management of STS. It can be used in advanced STS, either after failure of anthracyclines and ifosfamide or in patients unfit for these drugs, especially when reaching a high-tumor control and a long-term benefit is a priority. Toxicity profile is acceptable and manageable with no reported cumulative toxicities. Therefore, trabectedin has become one relevant therapeutic option in metastatic STS, especially in selected histologies.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dioxoles/therapeutic use , Sarcoma/diagnosis , Sarcoma/drug therapy , Tetrahydroisoquinolines/therapeutic use , Animals , Anthracyclines/pharmacokinetics , Anthracyclines/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Alkylating/pharmacokinetics , Clinical Trials as Topic/methods , Dioxoles/pharmacokinetics , Disease Management , Humans , Ifosfamide/pharmacokinetics , Ifosfamide/therapeutic use , Sarcoma/metabolism , Tetrahydroisoquinolines/pharmacokinetics , Trabectedin , Treatment Outcome
15.
Asia Pac J Clin Oncol ; 12(3): 259-64, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26948073

ABSTRACT

AIM: Limited sampling points for both amrubicin (AMR) and its active metabolite amrubicinol (AMR-OH) were simultaneously optimized using Akaike's information criterion (AIC) calculated by pharmacokinetic modeling. METHODS: In this pharmacokinetic study, 40 mg/m(2) of AMR was administered as a 5-min infusion on three consecutive days to 21 Japanese lung cancer patients. Blood samples were taken at 0, 0.08, 0.25, 0.5, 1, 2, 4, 8 and 24 h after drug infusion, and AMR and AMR-OH concentrations in plasma were quantitated using a high-performance liquid chromatography. The pharmacokinetic profile of AMR was characterized using a three-compartment model and that of AMR-OH using a one-compartment model following a first-order absorption process. These pharmacokinetic profiles were then integrated into one pharmacokinetic model for simultaneous fitting of AMR and AMR-OH. After fitting to the pharmacokinetic model, 65 combinations of four sampling points from the concentration profiles were evaluated for their AICs. Stepwise regression analysis was applied to select the sampling points for AMR and AMR-OH to predict the area under the concentration-time curves (AUCs) at best. RESULTS: Of the three combinations that yielded favorable AIC values, 0.25, 2, 4 and 8 h yielded the best AUC prediction for both AMR (R(2) = 0.977) and AMR-OH (R(2) = 0.886). The prediction error for AUC was less than 15%. CONCLUSION: The optimal limited sampling points of AMR and AMR-OH after AMR infusion were found to be 0.25, 2, 4 and 8 h, enabling less frequent blood sampling in further expanded pharmacokinetic studies for both AMR and AMR-OH.


Subject(s)
Anthracyclines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Lung Neoplasms/drug therapy , Aged , Area Under Curve , Female , Humans , Lung Neoplasms/metabolism , Male , Middle Aged
16.
Blood Rev ; 30(1): 55-64, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26321049

ABSTRACT

Acute myeloid leukemia is a clonal but heterogeneous disease differing in molecular pathogenesis, clinical features and response to chemotherapy. This latter frequently consists of a combination of cytarabine and anthracyclines, although etoposide, demethylating agents, and other drugs are also used. Unfortunately, chemoresistance is a common and serious problem. Multiple mechanisms account for impaired effectiveness of drugs and reduced levels of active agents in target cells. The latter can be due to lower drug uptake, increased export or decreased intracellular proportion of active/inactive agent due to changes in the expression/function of enzymes responsible for the activation of pro-drugs and the inactivation of active agents. Characterization of the "resistome", or profile of expressed genes accounting for multi-drug resistance (MDR) phenotype, would permit to predict the lack of response to chemotherapy and would help in the selection of the best pharmacological regime for each patient and moment, and to develop strategies of chemosensitization.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Anthracyclines/pharmacokinetics , Anthracyclines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Transport , Biomarkers, Pharmacological/metabolism , Biotransformation , Cytarabine/pharmacokinetics , Cytarabine/therapeutic use , Etoposide/pharmacokinetics , Etoposide/therapeutic use , Humans , Leukemia, Myeloid, Acute/pathology , Precision Medicine , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use
17.
Curr Drug Metab ; 16(6): 412-26, 2015.
Article in English | MEDLINE | ID: mdl-26321196

ABSTRACT

Many clinical studies involving anti-tumor agents neglect to consider how these agents are metabolized within the host and whether the creation of specific metabolites alters drug therapeutic properties or toxic side effects. However, this is not the case for the anthracycline class of chemotherapy drugs. This review describes the various enzymes involved in the one electron (semi-quinone) or two electron (hydroxylation) reduction of anthracyclines, or in their reductive deglycosidation into deoxyaglycones. The effects of these reductions on drug antitumor efficacy and toxic side effects are also discussed. Current evidence suggests that the one electron reduction of anthracyclines augments both their tumor toxicity and their toxicity towards the host, in particular their cardiotoxicity. In contrast, the two electron reduction (hydroxylation) of anthracyclines strongly reduces their ability to kill tumor cells, while augmenting cardiotoxicity through their accumulation within cardiomyocytes and their direct effects on excitation/contraction coupling within the myocytes. The reductive deglycosidation of anthracyclines appears to inactivate the drug and only occurs under rare, anaerobic conditions. This knowledge has resulted in the identification of important new approaches to improve the therapeutic index of anthracyclines, in particular by inhibiting their cardiotoxicity. The true utility of these approaches in the management of cancer patients undergoing anthracycline-based chemotherapy remains unclear, although one such agent (the iron chelator dexrazoxane) has recently been approved for clinical use.


Subject(s)
Anthracyclines/pharmacokinetics , Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Benzoquinones/metabolism , Humans , Hydroxylation , Oxidation-Reduction , Treatment Outcome
18.
Am J Clin Oncol ; 38(4): 405-10, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26214085

ABSTRACT

OBJECTIVES: We conducted a phase I trial of erlotinib, an epidermal growth factor receptor tyrosine kinase inhibitor, combined with amrubicin, a topoisomerase II inhibitor. The aim was to determine the maximum tolerated dose, the dose-limiting toxicities (DLTs), and the pharmacokinetics of this combination in patients with non-small cell lung cancer who had received previous chemotherapy. METHODS: A total of 9 patients with stage IV disease were treated at 3-week intervals with erlotinib once daily on days 1 through 21 plus a 5-minute intravenous injection of amrubicin on days 1 through 3. RESULTS: The dose levels evaluated were erlotinib (mg/body)/amrubicin (mg/m): 100/30 (n=3), 100/35 (n=3), and 150/30 (n=3). The maximum tolerated dose of erlotinib and amrubicin was 100 mg/body and 35 mg/m because 2 of the 3 patients experienced DLTs during the first cycle of treatment at the third dose level of 150 mg/body and 30 mg/m. Cessation of erlotinib administration for 8 days because of grade 3 leukopenia and grade 3 skin infection (erysipelas) were the DLTs. No drug-drug interactions between erlotinib and amrubicin were observed in this study. The overall response rate was 33%, including 3 partial responses, in the 9 patients. The median progression-free survival for all patients was quite long, 11.3 months, and the median overall survival has not yet been reached. CONCLUSIONS: Combined erlotinib plus amrubicin therapy seems to be highly effective, with acceptable toxicity, against non-small cell lung cancer. The recommended dose for phase II studies was erlotinib 100 mg once daily on days 1 through 21, and amrubicin 35 mg/m on days 1 through 3 administered every 21 days.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Anthracyclines/administration & dosage , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Erlotinib Hydrochloride , Erysipelas/chemically induced , Female , Humans , Leukopenia/chemically induced , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Quinazolines/administration & dosage , Quinazolines/adverse effects , Quinazolines/pharmacokinetics , Treatment Outcome
19.
Invest New Drugs ; 33(3): 594-602, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25698442

ABSTRACT

PURPOSE: 5-imino-13-deoxydoxorubicin (DIDOX; GPX-150) is a doxorubicin analog modified in two locations to prevent formation of cardiotoxic metabolites and reactive oxygen species. Preclinical studies have demonstrated anti-cancer activity without cardiotoxicity. A phase I study was performed in order to determine the maximum-tolerated dose (MTD) of GPX-150 in patients with metastatic solid tumors. METHODS: GPX-150 was administered as an intravenous infusion every 21 days for up to 8 cycles. An accelerated dose escalation was used for the first three treatment groups. The dosing groups were (A) 14 mg/m(2), (B) 28 mg/m(2), (C), 56 mg/m(2), (D) 84 mg/m(2), (E) 112 mg/m(2), (F) 150 mg/m(2), (G) 200 mg/m(2), and (H) 265 mg/m(2). Pharmacokinetic samples were drawn during the first 72 h of cycle 1. RESULTS: The MTD was considered to be reached at the highest dosing level of 265 mg/m(2) since dose reduction was required in 5 of 6 patients for neutropenia. The most frequent adverse events were neutropenia, anemia, fatigue, and nausea. No patients experienced cardiotoxicity while on the study. The best overall response was stable disease in four (20 %) patients. Pharmacokinetic analysis revealed an AUC of 8.0 (±2.6) µg · h/mL, a clearance of 607 (±210) mL/min/m(2) and a t1/2ß of 13.8 (±4.6) hours. CONCLUSIONS: GPX-150 administered every 21 days has an acceptable side effect profile and no cardiotoxicity was observed. Further investigation is needed to determine the efficacy of GPX-150 in anthracycline-sensitive malignancies.


Subject(s)
Anthracyclines/pharmacokinetics , Doxorubicin/analogs & derivatives , Neoplasms/drug therapy , Adult , Aged , Anthracyclines/chemistry , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Demography , Dose-Response Relationship, Drug , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , Humans , Male , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Stroke Volume/drug effects , Treatment Outcome
20.
Ann Hematol ; 93(8): 1381-90, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24994605

ABSTRACT

Glutathione S-transferases (GSTs) contribute to the metabolism of different xenobiotics and anticancer drugs and confer protection against oxidative stress thus may influence the treatment outcome of acute myeloid leukemia (AML). Studies regarding the association between GSTT1 and GSTM1 polymorphisms and treatment outcome in AML patients showed an inconsistent result. A systematic review and meta-analysis were performed to further explore this association. PubMed, Hartford User Group Exchange (HUGE), and China National Knowledge Infrastructure (CNKI) databases were searched for all related publications. Statistical analyses were analyzed by using RevMan 5.0 and Stata 9.0 softwares. A total of 1,837 patients in 11 studies were included. GSTT1 null genotype was found to be significantly associated with a reduced response after first course of induction chemotherapy (odds ratio (OR) = 0.894, 95 % confidence interval (CI) = 0.818-0.977, P = 0.013), progression-free survival (PFS; hazard ratio (HR) = 0.698, 95 % CI = 0.520-0.937, P = 0.017), and overall survival (OS; HR = 0.756, 95 % CI = 0.618-0.925, P = 0.007) in Asian population. GSTM1/GSTT1 double-null genotype was also identified to be significantly associated with response after the first course of induction chemotherapy (OR = 0.40, 95 % CI = 0.24-0.67, P = 0.0003). Our study suggested that GSTT1 null genotype and GSTT1/GSTM1 double-null genotype were associated with a worse treatment outcome for AML patients, especially in Asian population.


Subject(s)
Glutathione Transferase/genetics , Leukemia, Myeloid, Acute/genetics , Neoplasm Proteins/genetics , Adolescent , Adult , Aged , Anthracyclines/administration & dosage , Anthracyclines/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , China/epidemiology , Cytarabine/administration & dosage , Cytarabine/pharmacokinetics , Disease-Free Survival , Female , Gene Deletion , Genotype , Glutathione Transferase/analysis , Glutathione Transferase/physiology , Humans , Inactivation, Metabolic , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Mutation , Neoplasm Proteins/analysis , Neoplasm Proteins/physiology , Odds Ratio , Pharmacogenetics , Remission Induction , Survival Analysis , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...