1
|
Marini HR, Facchini BA, di Francia R, Freni J, Puzzolo D, Montella L, Facchini G, Ottaiano A, Berretta M, Minutoli L. Glutathione: Lights and Shadows in Cancer Patients. Biomedicines 2023; 11:2226. [PMID: 37626722 PMCID: PMC10452337 DOI: 10.3390/biomedicines11082226] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
In cases of cellular injury, there is an observed increase in the production of reactive oxygen species (ROS). When this production becomes excessive, it can result in various conditions, including cancerogenesis. Glutathione (GSH), the most abundant thiol-containing antioxidant, is fundamental to re-establishing redox homeostasis. In order to evaluate the role of GSH and its antioxi-dant effects in patients affected by cancer, we performed a thorough search on Medline and EMBASE databases for relevant clinical and/or preclinical studies, with particular regard to diet, toxicities, and pharmacological processes. The conjugation of GSH with xenobiotics, including anti-cancer drugs, can result in either of two effects: xenobiotics may lose their harmful effects, or GSH conjugation may enhance their toxicity by inducing bioactivation. While being an interesting weapon against chemotherapy-induced toxicities, GSH may also have a potential protective role for cancer cells. New studies are necessary to better explain the relationship between GSH and cancer. Although self-prescribed glutathione (GSH) implementation is prevalent among cancer patients with the intention of reducing the toxic effects of anticancer treatments and potentially preventing damage to normal tissues, this belief lacks substantial scientific evidence for its efficacy in reducing toxicity, except in the case of cisplatin-related neurotoxicity. Therefore, the use of GSH should only be considered under medical supervision, taking into account the appropriate timing and setting.
Collapse
Affiliation(s)
- Herbert Ryan Marini
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| | - Bianca Arianna Facchini
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80133 Napoli, Italy;
| | - Raffaele di Francia
- Gruppo Oncologico Ricercatori Italiani (GORI-ONLUS), 33170 Pordenone, Italy;
| | - José Freni
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (J.F.); (D.P.)
| | - Domenico Puzzolo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy; (J.F.); (D.P.)
| | - Liliana Montella
- Division of Medical Oncology, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, 80078 Pozzuoli, Italy; (L.M.); (G.F.)
| | - Gaetano Facchini
- Division of Medical Oncology, “Santa Maria delle Grazie” Hospital, ASL Napoli 2 Nord, 80078 Pozzuoli, Italy; (L.M.); (G.F.)
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, 80131 Napoli, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| | - Letteria Minutoli
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (H.R.M.); (L.M.)
| |
Collapse
|
2
|
Targeting Redox Regulation as a Therapeutic Opportunity against Acute Leukemia: Pro-Oxidant Strategy or Antioxidant Approach? Antioxidants (Basel) 2022; 11:antiox11091696. [PMID: 36139768 PMCID: PMC9495346 DOI: 10.3390/antiox11091696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/07/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
Redox adaptation is essential for human health, as the physiological quantities of non-radical reactive oxygen species operate as the main second messengers to regulate normal redox reactions by controlling several sensors. An abnormal increase reactive oxygen species, called oxidative stress, induces biological injury. For this reason, variations in oxidative stress continue to receive consideration as a possible approach to treat leukemic diseases. However, the intricacy of redox reactions and their effects might be a relevant obstacle; consequently, and alongside approaches aimed at increasing oxidative stress in neoplastic cells, antioxidant strategies have also been suggested for the same purpose. The present review focuses on the molecular processes of anomalous oxidative stress in acute myeloid and acute lymphoblastic leukemias as well as on the oxidative stress-determined pathways implicated in leukemogenic development. Furthermore, we review the effect of chemotherapies on oxidative stress and the possibility that their pharmacological effects might be increased by modifying the intracellular redox equilibrium through a pro-oxidant approach or an antioxidant strategy. Finally, we evaluated the prospect of varying oxidative stress as an efficacious modality to destroy chemoresistant cells using new methodologies. Altering redox conditions may be advantageous for inhibiting genomic variability and the eradication of leukemic clones will promote the treatment of leukemic disease.
Collapse
|
3
|
Verrilli AM, Leibman NF, Hohenhaus AE, Mosher BA. Safety and efficacy of a ribose-cysteine supplement to increase erythrocyte glutathione concentration in healthy dogs. Am J Vet Res 2021; 82:653-658. [PMID: 34296936 DOI: 10.2460/ajvr.82.8.653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the safety of oral administration of a d-ribose-l-cysteine (RibCys) supplement to dogs and the effect of this supplementation on erythrocyte glutathione (GSH) concentration. ANIMALS 24 healthy adult dogs. PROCEDURES In a randomized, double-blinded, controlled trial, dogs received 500 mg of a RibCys supplement or placebo (n = 12/group), PO, every 12 hours for 4 weeks. Dogs were evaluated weekly by means of a physical examination, CBC, serum biochemical analysis, urinalysis, and owner-completed quality-of-life questionnaire. Erythrocyte GSH concentration was measured on day 0 (ie, the day before treatment began) and weekly during supplementation. RESULTS No dose-limiting adverse effects were noted in any dog. Two dogs in each group had mild, self-limiting diarrhea and anemia. No significant increase in erythrocyte GSH concentration was noted in either group at any time point. Two dogs in the RibCys group had improved skin and coat health and improved clinical signs of osteoarthritis. No clinical or owner-perceived improvements were noted in the placebo group. CONCLUSIONS AND CLINICAL RELEVANCE The RibCys supplement was safe and well tolerated in all dogs. Owners reported improvements in dermatologic and orthopedic conditions in some dogs in the RibCys group. No significant differences were observed in erythrocyte GSH concentration before or after RibCys treatment. This lack of significant differences may have been attributable to the use of healthy dogs, which would not be expected to have depleted GSH concentrations. Given the observed safety profile of RibCys, additional research is warranted to explore the potential usefulness of RibCys supplementation in dogs with cancer and those undergoing treatment for cancer.
Collapse
|
4
|
Zou M, Du Y, Liu R, Zheng Z, Xu J. Nanocarrier-delivered small interfering RNA for chemoresistant ovarian cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 12:e1648. [PMID: 33682310 DOI: 10.1002/wrna.1648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 12/13/2022]
Abstract
Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States. Because success in early screening is limited, and most patients with advanced disease develop resistance to multiple treatment modalities, the overall prognosis of ovarian cancer is poor. Despite the revolutionary role of surgery and chemotherapy in curing ovarian cancer, recurrence remains a major challenge in treatment. Thus, improving our understanding of the pathogenesis of ovarian cancer is essential for developing more effective treatments. In this review, we analyze the underlying molecular mechanisms leading to chemotherapy resistance. We discuss the clinical benefits and potential challenges of using nanocarrier-delivered small interfering RNA to treat chemotherapy-resistant ovarian cancer. We aim to elicit collaborative studies on nanocarrier-delivered small interfering RNA to improve the long-term survival rate and quality of life of patients with ovarian cancer. This article is categorized under: RNA Methods > RNA Nanotechnology Regulatory RNAs/RNAi/Riboswitches > RNAi: Mechanisms of Action.
Collapse
Affiliation(s)
- Mingyuan Zou
- Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Yue Du
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruizhen Liu
- The First People's Hospital of Wu'an, Wu'an, Hebei, China
| | - Zeliang Zheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Jian Xu
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Research design considerations for chronic pain prevention clinical trials: IMMPACT recommendations. Pain Rep 2021; 6:e895. [PMID: 33981929 PMCID: PMC8108588 DOI: 10.1097/pr9.0000000000000895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 03/31/2015] [Accepted: 04/07/2015] [Indexed: 12/25/2022] Open
Abstract
Although certain risk factors can identify individuals who are most likely to develop chronic pain, few interventions to prevent chronic pain have been identified. To facilitate the identification of preventive interventions, an IMMPACT meeting was convened to discuss research design considerations for clinical trials investigating the prevention of chronic pain. We present general design considerations for prevention trials in populations that are at relatively high risk for developing chronic pain. Specific design considerations included subject identification, timing and duration of treatment, outcomes, timing of assessment, and adjusting for risk factors in the analyses. We provide a detailed examination of 4 models of chronic pain prevention (ie, chronic postsurgical pain, postherpetic neuralgia, chronic low back pain, and painful chemotherapy-induced peripheral neuropathy). The issues discussed can, in many instances, be extrapolated to other chronic pain conditions. These examples were selected because they are representative models of primary and secondary prevention, reflect persistent pain resulting from multiple insults (ie, surgery, viral infection, injury, and toxic or noxious element exposure), and are chronically painful conditions that are treated with a range of interventions. Improvements in the design of chronic pain prevention trials could improve assay sensitivity and thus accelerate the identification of efficacious interventions. Such interventions would have the potential to reduce the prevalence of chronic pain in the population. Additionally, standardization of outcomes in prevention clinical trials will facilitate meta-analyses and systematic reviews and improve detection of preventive strategies emerging from clinical trials.
Collapse
|
6
|
Mahmod II, Ismail IS, Alitheen NB, Normi YM, Abas F, Khatib A, Rudiyanto, Latip J. NMR and LCMS analytical platforms exhibited the nephroprotective effect of Clinacanthus nutans in cisplatin-induced nephrotoxicity in the in vitro condition. BMC Complement Med Ther 2020; 20:320. [PMID: 33092571 PMCID: PMC7579835 DOI: 10.1186/s12906-020-03067-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 09/01/2020] [Indexed: 12/29/2022] Open
Abstract
Background Clinacanthus nutans (C. nutans) Lind. locally known as Belalai Gajah or Sabah snake grass is a medicinal plant belonging to Acanthaceae family. In Asia, this plant is traditionally used for treating skin rashes, insects and snake bites, diabetes mellitus, fever and for diuretic effect. C. nutans has been reported to possess biological activities including anti-oxidant, anti-inflammation, anti-cancer, anti-diabetic and anti-viral activities. Methods Proton Nuclear Magnetic Resonance (1H NMR) and Liquid Chromatography Mass Spectroscopy (LCMS) coupled with multivariate data analysis were employed to characterize the metabolic variations of intracellular metabolites and the compositional changes of the corresponding culture media in rat renal proximal tubular cells (NRK-52E). Results NMR and LCMS analysis highlighted choline, creatine, phosphocholine, valine, acetic acid, phenylalanine, leucine, glutamic acid, threonine, uridine and proline as the main metabolites which differentiated the cisplatin-induced group of NRK-52E from control cells extract. The corresponding media exhibited lactic acid, glutamine, glutamic acid and glucose-1-phosphate as the varied metabolites. The altered pathways perturbed by cisplatin nephrotoxic on NRK-52E cells included changes in amino acid metabolism, lipid metabolism and glycolysis. Conclusion The C. nutans aqueous extract (1000 μg/mL) exhibited the most potential nephroprotective effect against cisplatin toxicity on NRK-52E cell lines at 89% of viability. The protective effect could be seen through the changes of the metabolites such as choline, alanine and valine in the C. nutans pre-treated samples with those of the cisplatin-induced group. Supplementary information Supplementary information accompanies this paper at 10.1186/s12906-020-03067-3.
Collapse
Affiliation(s)
- Ilya Iryani Mahmod
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Intan Safinar Ismail
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia. .,Department of Chemistry, Faculty of Science, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Yahaya M Normi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Faridah Abas
- Laboratory of Natural Products, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.,Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Alfi Khatib
- Faculty of Pharmacy, International Islamic University Malaysia, 25200, Kuantan, Pahang, Malaysia
| | - Rudiyanto
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, 21030, Kuala Nerus, Terengganu, Malaysia
| | - Jalifah Latip
- School of Chemical Science and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, Bandar Baru Bangi, Selangor, Malaysia
| |
Collapse
|
7
|
Protective effect of the oral administration of cystine and theanine on oxaliplatin-induced peripheral neuropathy: a pilot randomized trial. Int J Clin Oncol 2020; 25:1814-1821. [PMID: 32594273 PMCID: PMC7498479 DOI: 10.1007/s10147-020-01728-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 06/15/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Oxaliplatin, one of the key cytotoxic drugs for colorectal cancer, frequently causes peripheral neuropathy which leads to dose modification and decreased patients' quality of life. However, prophylactic or therapeutic measures have not yet been established. Orally administered amino acids, cystine and theanine, promoted the synthesis of glutathione which was one of the potential candidates for preventing the neuropathy. The aim of this study was to determine whether daily oral administration of cystine and theanine attenuated oxaliplatin-induced peripheral neuropathy (OXLIPN). METHODS Twenty-eight colorectal cancer patients who received infusional 5-fluorouracil, leucovorin, and oxaliplatin (mFOLFOX6) therapy were randomly and evenly assigned to the cystine and theanine group and the control group. OXLIPN was assessed up to the sixth course using original 7-item questionnaire as well as Common Terminology Criteria for Adverse Events (CTCAE) grading scale. RESULTS Neuropathy scores according to our original questionnaire were significantly smaller in the cystine and theanine group at the fourth (p = 0.026), fifth (p = 0.029), and sixth course (p = 0.038). Furthermore, significant differences were also observed in CTCAE neuropathy grades at the fourth (p = 0.037) and the sixth course (p = 0.017). There was one patient in each group who required dose reduction due to OXLIPN. Except for neurotoxicity, no significant differences were noted in the incidence of adverse events, and the total amount of administered oxaliplatin. CONCLUSION The results demonstrated the daily oral administration of cystine and theanine attenuated OXLIPN.
Collapse
|
8
|
Staff NP, Cavaletti G, Islam B, Lustberg M, Psimaras D, Tamburin S. Platinum-induced peripheral neurotoxicity: From pathogenesis to treatment. J Peripher Nerv Syst 2020; 24 Suppl 2:S26-S39. [PMID: 31647151 DOI: 10.1111/jns.12335] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022]
Abstract
Platinum-induced peripheral neurotoxicity (PIPN) is a common side effect of platinum-based chemotherapy that may cause dose reduction and discontinuation, with oxaliplatin being more neurotoxic. PIPN includes acute neurotoxicity restricted to oxaliplatin, and chronic non-length-dependent sensory neuronopathy with positive and negative sensory symptoms and neuropathic pain in both upper and lower limbs. Chronic sensory axonal neuropathy manifesting as stocking-and-glove distribution is also frequent. Worsening of neuropathic symptoms after completing the last chemotherapy course may occur. Motor and autonomic involvement is uncommon. Ototoxicity is frequent in children and more commonly to cisplatin. Platinum-based compounds result in more prolonged neuropathic symptoms in comparison to other chemotherapy agents. Patient reported outcomes questionnaires, clinical evaluation and instrumental tools offer complementary information in PIPN. Electrodiagnostic features include diffusely reduced/abolished sensory action potentials, in keeping with a sensory neuronopathy. PIPN is dependent on cumulative dose but there is a large variability in its occurrence. The search for additional risk factors for PIPN has thus far yielded no consistent findings. There are currently no neuroprotective strategies to reduce the risk of PIPN, and symptomatic treatment is limited to duloxetine that was found effective in a single phase III intervention study. This review critically examines the pathogenesis, incidence, risk factors (both clinical and pharmacogenetic), clinical phenotype and management of PIPN.
Collapse
Affiliation(s)
- Nathan P Staff
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Guido Cavaletti
- Experimental Neurology Unit, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Badrul Islam
- Laboratory Sciences and Services Division, The International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Maryam Lustberg
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Dimitri Psimaras
- OncoNeuroTox Group, Center for Patients with Neurological Complications of Oncologic Treatments, Hôpitaux Universitaires Pitié-Salpetrière-Charles Foix et Hôpital Percy, Paris, France
| | - Stefano Tamburin
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| |
Collapse
|
9
|
Hamroun A, Lenain R, Bigna JJ, Speyer E, Bui L, Chamley P, Pottier N, Cauffiez C, Dewaeles E, Dhalluin X, Scherpereel A, Hazzan M, Maanaoui M, Glowacki F. Prevention of Cisplatin-Induced Acute Kidney Injury: A Systematic Review and Meta-Analysis. Drugs 2020; 79:1567-1582. [PMID: 31429065 DOI: 10.1007/s40265-019-01182-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE Cisplatin-induced acute kidney injury (CIA) is a serious adverse event that affects 20-40% of exposed patients, despite any implemented precaution to avoid it. The aim of this work was therefore to identify a relevant nephroprotective method for CIA. METHODS We searched Pubmed, Embase, and Web of Science from 1 January 1978 to 1 June 2018, without language restriction. All studies (observational and interventional) assessing a CIA prevention method for adults receiving at least one course of cisplatin were eligible. The primary outcome was acute nephrotoxicity, as defined by the AKI-KDIGO classification (2012). The odds ratio and corresponding 95% confidence interval were used to assess the associations. We used narrative synthesis in case of heterogeneity regarding intervention, population, or outcome. When possible, a random-effects model was used to pool studies. The heterogeneity between studies was quantified (I2), and multiple meta-regressions were carried out to identify potential confounders. RESULTS Within 4520 eligible studies, 51 articles fulfilling the selection criteria were included in the review, assessing 21 different prevention methods. A meta-analysis could only be performed on the 15 observational studies concerning magnesium supplementation (1841 patients), and showed a significant nephroprotective effect for all combined grades of CIA (OR 0.24, [0.19-0.32], I2 = 0.0%). This significant nephroprotective effect was also observed for grades 2 and 3 CIA (OR 0.22, [0.14-0.33], I2 = 0.0% and OR 0.25, [0.08-0.76], I2 = 0.0%, respectively). CONCLUSION While no method of prevention had so far demonstrated its indisputable efficacy, our results highlight the potential protective effect of magnesium supplementation on cisplatin-induced acute nephrotoxicity. TRIAL REGISTRATION This study is registered in PROSPERO, CRD42018090612.
Collapse
Affiliation(s)
- Aghilès Hamroun
- Nephrology Department, CHRU Lille, University of Lille, 59000, Lille, France.
| | - Rémi Lenain
- Nephrology Department, CHRU Lille, University of Lille, 59000, Lille, France
| | - Jean Joel Bigna
- Faculty of Medicine, University of Paris Saclay, Paris, France
| | - Elodie Speyer
- Centre for Research in Epidemiology and Population Health (CESP), Paris Saclay University, Paris Sud University, Versailles Saint Quentin University, INSERM UMRS 1018, 94807, Villejuif, France
| | - Linh Bui
- Nephrology Department, CH Beuvry, Béthune, France
| | - Paul Chamley
- Nephrology Department, CHRU Lille, University of Lille, 59000, Lille, France
| | - Nicolas Pottier
- Department of Toxicology and Genetic Pathologies, CHRU Lille, 59000, Lille, France
| | - Christelle Cauffiez
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Medicine Faculty, Research Department, University of Lille, 59045, Lille, France
| | - Edmone Dewaeles
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Medicine Faculty, Research Department, University of Lille, 59045, Lille, France
| | - Xavier Dhalluin
- Pulmonary and Thoracic Oncology Department, University of Lille, INSERM U1189 OncoThAI, 59000, Lille, France
| | - Arnaud Scherpereel
- Pulmonary and Thoracic Oncology Department, University of Lille, INSERM U1189 OncoThAI, 59000, Lille, France
| | - Marc Hazzan
- Nephrology Department, CHRU Lille, University of Lille, 59000, Lille, France
- INSERM, UMR995, 59000, Lille, France
| | - Mehdi Maanaoui
- Nephrology Department, CHRU Lille, University of Lille, 59000, Lille, France
| | - François Glowacki
- Nephrology Department, CHRU Lille, University of Lille, 59000, Lille, France
- EA 4483-IMPECS-IMPact of Environmental ChemicalS on Human Health, Medicine Faculty, Research Department, University of Lille, 59045, Lille, France
| |
Collapse
|
10
|
Santos NAGD, Ferreira RS, Santos ACD. Overview of cisplatin-induced neurotoxicity and ototoxicity, and the protective agents. Food Chem Toxicol 2019; 136:111079. [PMID: 31891754 DOI: 10.1016/j.fct.2019.111079] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Cisplatin has dramatically improved the survival rate of cancer patients, but it has also increased the prevalence of hearing and neurological deficits in this population. Cisplatin induces ototoxicity, peripheral (most prevalent) and central (rare) neurotoxicity. This review addresses the ototoxicity and the neurotoxicity associated with cisplatin-based chemotherapy, providing an integrated view of the potential protective agents that have been evaluated in vitro, in vivo and in clinical trials, their targets and mechanisms of protection and their effects on the antitumor activity of cisplatin. So far, the findings are insufficient to support the use of any oto- or neuroprotective agent before, during or after cisplatin chemotherapy. Despite their promising effects in vitro and in animal studies, many agents have not been evaluated in clinical trials. Additionally, the clinical trials have limitations concerning the sample size, controls, measurement, heterogeneous groups, several arms of treatment, short follow-up or no blinding. Besides that, for most agents, the effects on the antitumor activity of cisplatin have not been evaluated in tumor-bearing animals, which discourages clinical trials. Further well-designed randomized controlled clinical trials are necessary to definitely demonstrate the effectiveness of the oto- or neuroprotective agents proposed by animal and in vitro studies.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Cardozo Dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
11
|
α-Lipoic acid prevents against cisplatin cytotoxicity via activation of the NRF2/HO-1 antioxidant pathway. PLoS One 2019; 14:e0226769. [PMID: 31877176 PMCID: PMC6932784 DOI: 10.1371/journal.pone.0226769] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/03/2019] [Indexed: 12/30/2022] Open
Abstract
The production of reactive oxygen species (ROS) by cisplatin is one of the major mechanisms of cisplatin-induced cytotoxicity. We examined the preventive effect of α-lipoic acid (LA) on cisplatin-induced toxicity via its antioxidant effects on in vitro and ex vivo culture systems. To elucidate the mechanism of the antioxidant activity of LA, NRF2 was inhibited using NRF2 siRNA, and the change in antioxidant activity of LA was characterized. MTT assays showed that LA was safe at concentrations up to 0.5 mM in HEI-OC1 cells and had a protective effect against cisplatin-induced cytotoxicity. Intracellular ROS production in HEI-OC1 cells was rapidly increased by cisplatin for up to 48 h. However, treatment with LA significantly reduced the production of ROS and increased the expression of the antioxidant proteins HO-1 and SOD1. Ex vivo, the organs of Corti of the group pretreated with LA exhibited better preservation than the group that received cisplatin alone. We also confirmed the nuclear translocation of NRF2 after LA administration, and that NRF2 inhibition decreased the antioxidant activity of LA. Together, these results indicate that the antioxidant activity of LA was through the activation of the NRF2/HO-1 antioxidant pathway.
Collapse
|
12
|
Bhuyan DJ, Alsherbiny MA, Perera S, Low M, Basu A, Devi OA, Barooah MS, Li CG, Papoutsis K. The Odyssey of Bioactive Compounds in Avocado ( Persea americana) and Their Health Benefits. Antioxidants (Basel) 2019; 8:E426. [PMID: 31554332 PMCID: PMC6826385 DOI: 10.3390/antiox8100426] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 09/22/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022] Open
Abstract
Persea americana, commonly known as avocado, has recently gained substantial popularity and is often marketed as a "superfood" because of its unique nutritional composition, antioxidant content, and biochemical profile. However, the term "superfood" can be vague and misleading, as it is often associated with unrealistic health claims. This review draws a comprehensive summary and assessment of research performed in the last few decades to understand the nutritional and therapeutic properties of avocado and its bioactive compounds. In particular, studies reporting the major metabolites of avocado, their antioxidant as well as bioavailability and pharmacokinetic properties, are summarized and assessed. Furthermore, the potential of avocado in novel drug discovery for the prevention and treatment of cancer, microbial, inflammatory, diabetes, and cardiovascular diseases is highlighted. This review also proposes several interesting future directions for avocado research.
Collapse
Affiliation(s)
- Deep Jyoti Bhuyan
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Muhammad A Alsherbiny
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia.
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Saumya Perera
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Mitchell Low
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Amrita Basu
- Research Centre for Toxic Compounds in the Environment, Masaryk University, Brno 62500, Czech.
| | - Okram Abemsana Devi
- Department of Food Science and Nutrition, College of Community Science, Assam Agricultural University, Assam 785013, India.
| | - Mridula Saikia Barooah
- Department of Food Science and Nutrition, College of Community Science, Assam Agricultural University, Assam 785013, India.
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Konstantinos Papoutsis
- School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
13
|
Efficacy of Antioxidant Supplements on Prevention and Amelioration of Cisplatin-Induced Nephrotoxicity: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Jundishapur J Nat Pharm Prod 2019. [DOI: 10.5812/jjnpp.61527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
14
|
Singh N, Nayak J, Sahoo SK, Kumar R. Glutathione conjugated superparamagnetic Fe3O4-Au core shell nanoparticles for pH controlled release of DOX. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 100:453-465. [DOI: 10.1016/j.msec.2019.03.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/12/2019] [Accepted: 03/09/2019] [Indexed: 10/27/2022]
|
15
|
Chan S, Wang R, Man K, Nicholls J, Li H, Sun H, Chan GCF. A Novel Synthetic Compound, Bismuth Zinc Citrate, Could Potentially Reduce Cisplatin-Induced Toxicity Without Compromising the Anticancer Effect Through Enhanced Expression of Antioxidant Protein. Transl Oncol 2019; 12:788-799. [PMID: 30921749 PMCID: PMC6438849 DOI: 10.1016/j.tranon.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 02/14/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
Cisplatin is a common anticancer drug, but it comes with significant nephrotoxicity. Further cisplatin-induced oxidative stress contributes to the pathogenesis of the nephrotoxicity. A new compound, BiZn, can potentially prevent this complication. We verified our postulation by in vitro and in vivo models. From our findings, BiZn did not affect cisplatin-induced cytotoxicity on neuroblastoma cells under both in vitro and in vivo settings. However, BiZn significantly reduced the blood urea nitrogen and creatinine levels in cisplatin-treated mice. Under the lethal dosage of cisplatin, co-treatment of BiZn significantly increased the survival rate. BiZn stimulated antioxidant proteins metallothionein (MT) and glutathione (GSH) generation from kidney cells and minimized cisplatin-induced apoptosis. Knocking down MT-IIA and inhibiting GSH abolished such protection. In conclusion, pretreatment of BiZn decreased cisplatin-induced renal toxicity without affecting its antitumor activity. BiZn-induced antioxidant proteins MT and GSH may contribute to the renal protection effect.
Collapse
Affiliation(s)
- Shing Chan
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong
| | - Runming Wang
- Department of Chemistry, Faculty of Science, The University of Hong Kong
| | - Kwan Man
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong
| | - John Nicholls
- Department of Pathology, LKS Faculty of Medicine, The University of Hong Kong
| | - Hongyan Li
- Department of Chemistry, Faculty of Science, The University of Hong Kong
| | - Hongzhe Sun
- Department of Chemistry, Faculty of Science, The University of Hong Kong.
| | - Godfrey Chi-Fung Chan
- Department of Paediatrics & Adolescent Medicine, LKS Faculty of Medicine, The University of Hong Kong.
| |
Collapse
|
16
|
Lee D, Kanzawa-Lee G, Knoerl R, Wyatt G, Smith EML. Characterization of Internal Validity Threats to Phase III Clinical Trials for Chemotherapy-Induced Peripheral Neuropathy Management: A Systematic Review. Asia Pac J Oncol Nurs 2019; 6:318-332. [PMID: 31572750 PMCID: PMC6696803 DOI: 10.4103/apjon.apjon_14_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective: The recent American Society of Clinical Oncology (ASCO) Clinical Guidelines for chemotherapy-induced peripheral neuropathy (CIPN) management (48 Phase III trials reviewed) only recommend duloxetine. However, before concluding that a CIPN intervention is ineffective, scientists and clinicians should consider the risk of Type II error in Phase III studies. The purpose of this systematic review was to characterize internal threats to validity in Phase III CIPN management trials. Methods: The PubMed, CINAHL, EMBASE®, and Scopus databases were searched for Phase III clinical trials testing interventions for CIPN management between 1990 and 2018. The key search terms were neoplasms, cancer, neuropathy, and CIPN. Two independent researchers evaluated 24 studies, using a modified Joanna Briggs Institute Checklist for Randomized Control Trials developed by the authors specific for CIPN intervention trials. Results: Two studies exhibited minimal or no design flaws. 22/24 Phase III clinical trials for CIPN have two or greater design flaws due to sample heterogeneity, malapropos mechanism of action, malapropos intervention dose, malapropos timing of the outcome measurement, confounding variables, lack of a valid and reliable measurement, and suboptimal statistical validity. Conclusions: Numerous CIPN interventions have been declared ineffective based on the results of Phase III trials. However, internal validity threats to numerous studies may have resulted in Type II error and subsequent dismissal of a potentially effective intervention. Patients may benefit from rigorous retesting of several agents (e.g., alpha-lipoic acid, duloxetine, gabapentin, glutathione, goshajinkigan, lamotrigine, nortriptyline, venlafaxine, and Vitamin E) to expand and validate the evidence regarding ASCO's recommendations for CIPN management.
Collapse
Affiliation(s)
- Deborah Lee
- Michigan State University, School of Nursing, East Lansing, Ann Arbor, MI, USA
| | | | - Robert Knoerl
- Phyllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gwen Wyatt
- Michigan State University, School of Nursing, East Lansing, Ann Arbor, MI, USA
| | | |
Collapse
|
17
|
Smith EML, Knoerl R, Yang JJ, Kanzawa-Lee G, Lee D, Bridges CM. In Search of a Gold Standard Patient-Reported Outcome Measure for Use in Chemotherapy- Induced Peripheral Neuropathy Clinical Trials. Cancer Control 2018; 25:1073274818756608. [PMID: 29480026 PMCID: PMC5925747 DOI: 10.1177/1073274818756608] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose: To test a reduced version—CIPN15—of the European Organisation for Research and Treatment of Cancer (EORTC) Quality of Life Questionnaire Chemotherapy-Induced Peripheral Neuropathy scale (QLQ-CIPN20) to establish a possible gold-standard patient-reported outcome measure for chemotherapy-induced peripheral neuropathy (CIPN). Methods: Using a prospective, longitudinal, case–control design, patients (n = 121) receiving neurotoxic chemotherapy completed the CIPN15 at baseline and 12 weeks and underwent objective neurological assessment using the 5-item Total Neuropathy Score-Clinical (TNSc). Healthy controls (n = 30) completed the CIPN15 once. Structural validity was evaluated using factor analysis. Because a stable factor structure was not found, a sum score was used to evaluate measures of the CIPN15’s psychometric properties—reliability, validity, sensitivity, and responsiveness—as follows: internal consistency via Cronbach’s α and item–item correlations; test–retest reliability via correlation between 2 CIPN15 scores from each patient; concurrent validity via correlation between CIPN15 and 5-item TNSc scores; contrasting group validity via comparison of CIPN15 scores from patients and healthy controls; sensitivity via descriptive statistics (means, standard deviation, ranges); and responsiveness via Cohen’s d effect size. Results: Most patients received single agent oxaliplatin (33.7%), paclitaxel (21.2%), or more than 1 neurotoxic drug concurrently (29.8%). Factor analysis revealed no stable factor structure. Cronbach’s α for the CIPN15 sum score was 0.91 (confidence interval [CI] = 0.89-0.93). Test–retest reliability was demonstrated based on strong correlations between the 2 scores obtained at the 12-week time point (r = 0.86; CI = 0.80-0.90). The CIPN15 and 5-item TNSc items reflecting symptoms (not signs) were moderately correlated (r range 0.57-0.72): concurrent validity. Statistically significant differences were found between patient and healthy control CIPN15 mean scores (P < .0001): contrasting group validity. All items encompassed the full score range but the CIPN15 linearly converted sum score did not: sensitivity. The CIPN15 was responsive based on a Cohen’s d of 0.52 (CI = 0.25-0.79). Conclusion: The sum-scored CIPN15 is reliable, valid, sensitive, and responsive when used to assess taxane- and platinum-induced CIPN.
Collapse
Affiliation(s)
| | - Robert Knoerl
- 2 Phylllis F. Cantor Center for Research in Nursing and Patient Care Services, Dana Farber Cancer Institute, Boston, MA, USA
| | - James J Yang
- 1 University of Michigan School of Nursing, Ann Arbor, MI, USA
| | | | - Deborah Lee
- 1 University of Michigan School of Nursing, Ann Arbor, MI, USA
| | - Celia M Bridges
- 1 University of Michigan School of Nursing, Ann Arbor, MI, USA
| |
Collapse
|
18
|
El-shafaei A, Abdelmaksoud R, Elshorbagy A, Zahran N, Elabd R. Protective effect of melatonin versus montelukast in cisplatin-induced seminiferous tubule damage in rats. Andrologia 2018; 50:e13077. [DOI: 10.1111/and.13077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Adel El-shafaei
- Department of Dermatology, Venereology and Andrology; Faculty of Medicine; Alexandria University; Alexandria Egypt
| | - Rania Abdelmaksoud
- Department of Dermatology, Venereology and Andrology; Faculty of Medicine; Alexandria University; Alexandria Egypt
| | - Amany Elshorbagy
- Department of Physiology; Faculty of Medicine; Alexandria University; Alexandria Egypt
| | - Noha Zahran
- Department of Histology and Cell biology; Faculty of Medicine; Alexandria University; Alexandria Egypt
| | - Rana Elabd
- Department of Dermatology, Venereology and Andrology; Faculty of Medicine; Alexandria University; Alexandria Egypt
| |
Collapse
|
19
|
Nunes SC, Serpa J. Glutathione in Ovarian Cancer: A Double-Edged Sword. Int J Mol Sci 2018; 19:ijms19071882. [PMID: 29949936 PMCID: PMC6073569 DOI: 10.3390/ijms19071882] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 01/21/2023] Open
Abstract
Glutathione (GSH) has several roles in a cell, such as a reactive oxygen species (ROS) scavenger, an intervenient in xenobiotics metabolism and a reservoir of cysteine. All of these activities are important in the maintenance of normal cells homeostasis but can also constitute an advantage for cancer cells, allowing disease progression and resistance to therapy. Ovarian cancer is the major cause of death from gynaecologic disease and the second most common gynaecologic malignancy worldwide. In over 50 years, the overall survival of patients diagnosed with epithelial ovarian cancer has not changed, regardless of the efforts concerning early detection, radical surgery and new therapeutic approaches. Late diagnosis and resistance to therapy are the main causes of this outcome, and GSH is profoundly associated with chemoresistance to platinum salts, which, together with taxane-based chemotherapy and surgery, are the main therapy strategies in ovarian cancer treatment. Herein, we present some insights into the role of GSH in the poor prognosis of ovarian cancer, and also point out how some strategies underlying the dependence of ovarian cancer cells on GSH can be further used to improve the effectiveness of therapy.
Collapse
Affiliation(s)
- Sofia C Nunes
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal.
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto, 1099-023 Lisboa, Portugal.
| | - Jacinta Serpa
- Centro de Estudos de Doenças Crónicas (CEDOC), NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisboa, Portugal.
- Unidade de Investigação em Patobiologia Molecular do Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof. Lima Basto, 1099-023 Lisboa, Portugal.
| |
Collapse
|
20
|
Pharmacometabolomics reveals a role for histidine, phenylalanine, and threonine in the development of paclitaxel-induced peripheral neuropathy. Breast Cancer Res Treat 2018; 171:657-666. [PMID: 29946863 DOI: 10.1007/s10549-018-4862-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023]
Abstract
PURPOSE Approximately 25% of breast cancer patients experience treatment delays or discontinuation due to paclitaxel-induced peripheral neuropathy (PN). Currently, there are no predictive biomarkers of PN. Pharmacometabolomics is an informative tool for biomarker discovery of drug toxicity. We conducted a secondary whole blood pharmacometabolomics analysis to assess the association between pretreatment metabolome, early treatment-induced metabolic changes, and the development of PN. METHODS Whole blood samples were collected pre-treatment (BL), just before the end of the first paclitaxel infusion (EOI), and 24 h after the first infusion (24H) from sixty patients with breast cancer receiving (80 mg/m2) weekly treatment. Neuropathy was assessed at BL and prior to each infusion using the sensory subscale (CIPN8) of the EORTC CIPN20 questionnaire. Blood metabolites were quantified from 1-D-1H-nuclear magnetic resonance spectra using Chenomx® software. Metabolite concentrations were normalized in preparation for Pearson correlation and one-way repeated measures ANOVA with multiple comparisons corrected by false discovery rate (FDR). RESULTS Pretreatment histidine, phenylalanine, and threonine concentrations were inversely associated with maximum change in CIPN8 (ΔCIPN8) (p < 0.02; FDR ≤ 25%). Paclitaxel caused a significant change in concentrations of 2-hydroxybutyrate, 3-hydroxybutyrate, pyruvate, o-acetylcarnitine, and several amino acids from BL to EOI and/or 24H (p < 0.05; FDR ≤ 25%), although these changes were not associated with ΔCIPN8. CONCLUSIONS Whole blood metabolomics is a feasible approach to identify potential biomarker candidates of paclitaxel-induced PN. The findings suggest that pretreatment concentrations of histidine, phenylalanine, and threonine may be predictive of the severity of future PN and paclitaxel-induced metabolic changes may be related to disruption of energy homeostasis.
Collapse
|
21
|
Potnuri AG, Allakonda L, Lahkar M. Crocin attenuates cyclophosphamide induced testicular toxicity by preserving glutathione redox system. Biomed Pharmacother 2018; 101:174-180. [DOI: 10.1016/j.biopha.2018.02.068] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 01/26/2023] Open
|
22
|
Antioxidants as precision weapons in war against cancer chemotherapy induced toxicity - Exploring the armoury of obscurity. Saudi Pharm J 2017; 26:177-190. [PMID: 30166914 PMCID: PMC6111235 DOI: 10.1016/j.jsps.2017.12.013] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 12/14/2017] [Indexed: 12/14/2022] Open
Abstract
Cancer is the leading cause of mortality worldwide, accounting for almost 13% of deaths in the world. Among the conventional cancer treatments, chemotherapy is most frequently carried out to treat malignant cancer rather than localised lesions which is amenable to surgery and radiotherapy. However, anticancer drugs are associated with a plethora of side effects. Each drug, within every class, has its own set of adverse reactions which may cause patient incompliance and deterioration of the quality of life. One of the major causes of adverse reactions, especially for drugs targeting DNA, is the excessive production of reactive oxygen species (ROS) and subsequent build up of oxidative stress. To curb these undesired side effects, several dietary supplements have been tested, amongst which antioxidants have gained increasing popularity as adjuvant in chemotherapy. However, many oncologists discourage the use of antioxidant rich food supplements because these may interfere with the modalities which kill cancer by generating free radicals. In the present review, all studies reporting concomitant use of several antioxidants with chemotherapy are indiscriminately included and discussed impartially. The effect of supplementation of thirteen different antioxidants and their analogues as a single agent or in combination with chemotherapy has been compiled in this article. The present review encompasses a total of 174 peer-reviewed original articles from 1967 till date comprising 93 clinical trials with a cumulative number of 18,208 patients, 56 animal studies and 35 in vitro studies. Our comprehensive data suggests that antioxidant has superior potential of ameliorating chemotherapeutic induced toxicity. Antioxidant supplementation during chemotherapy also promises higher therapeutic efficiency and increased survival times in patients.
Collapse
|
23
|
Bakogeorgos M, Georgoulias V. Risk-reduction and treatment of chemotherapy-induced peripheral neuropathy. Expert Rev Anticancer Ther 2017; 17:1045-1060. [PMID: 28868935 DOI: 10.1080/14737140.2017.1374856] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Chemotherapy-induced peripheral neuropathy (CIPN), a common adverse effect of several chemotherapeutic agents, has a significant impact on quality of life and may even compromise treatment efficacy, requiring chemotherapy dose reduction or discontinuation. CIPN is predominantly related with sensory rather than motor symptoms and the most common related cytotoxic agents are platinum compounds, taxanes and vinca alkaloids. CIPN symptoms may resolve after treatment cessation, but they can also be permanent and continue for years. Areas covered: We present an overview of CIPN pathophysiology, clinical assessment, prevention and treatment identified through a Pubmed search. Expert commentary: No substantial progress has been made in the last few years within the field of prevention and/or treatment of CIPN, in spite of remarkable efforts. Continuous research could expand our knowledge about chemotherapeutic-specific neuropathic pathways and eventually lead to the conception of innovative and targeted agents for the prevention and/or treatment of this debilitating chemotherapy adverse effect.
Collapse
|
24
|
Little AC, Sulovari A, Danyal K, Heppner DE, Seward DJ, van der Vliet A. Paradoxical roles of dual oxidases in cancer biology. Free Radic Biol Med 2017; 110:117-132. [PMID: 28578013 PMCID: PMC5535817 DOI: 10.1016/j.freeradbiomed.2017.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
Dysregulated oxidative metabolism is a well-recognized aspect of cancer biology, and many therapeutic strategies are based on targeting cancers by altering cellular redox pathways. The NADPH oxidases (NOXes) present an important enzymatic source of biological oxidants, and the expression and activation of several NOX isoforms are frequently dysregulated in many cancers. Cell-based studies have demonstrated a role for several NOX isozymes in controlling cell proliferation and/or cell migration, further supporting a potential contributing role for NOX in promoting cancer. While various NOX isoforms are often upregulated in cancers, paradoxical recent findings indicate that dual oxidases (DUOXes), normally prominently expressed in epithelial lineages, are frequently suppressed in epithelial-derived cancers by epigenetic mechanisms, although the functional relevance of such DUOX silencing has remained unclear. This review will briefly summarize our current understanding regarding the importance of reactive oxygen species (ROS) and NOXes in cancer biology, and focus on recent observations indicating the unique and seemingly opposing roles of DUOX enzymes in cancer biology. We will discuss current knowledge regarding the functional properties of DUOX, and recent studies highlighting mechanistic consequences of DUOX1 loss in lung cancer, and its consequences for tumor invasiveness and current anticancer therapy. Finally, we will also discuss potentially unique roles for the DUOX maturation factors. Overall, a better understanding of mechanisms that regulate DUOX and the functional consequences of DUOX silencing in cancer may offer valuable new diagnostic insights and novel therapeutic opportunities.
Collapse
Affiliation(s)
- Andrew C Little
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States
| | - Arvis Sulovari
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States; Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David J Seward
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States.
| |
Collapse
|
25
|
Fu X, Wu H, Li J, Wang C, Li M, Ma Q, Yang W. Efficacy of Drug Interventions for Chemotherapy-Induced Chronic Peripheral Neurotoxicity: A Network Meta-analysis. Front Neurol 2017. [PMID: 28642731 PMCID: PMC5462987 DOI: 10.3389/fneur.2017.00223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Peripheral neurotoxicity is a disturbing issue for cancer patients who are treated with chemotherapy. Several medications have been developed for preventing chemotherapy-induced chronic neurotoxicity (CICNT) however; their relative efficacies have not yet been studied. In this study, we conducted a network meta-analysis to give intervention recommendations. The literature was searched in a variety of databases and eligible studies were chosen based on predefined criteria. Data extraction and statistical analysis was performed, and the results are displayed using the odds ratio (OR) and corresponding 95% credible intervals (CrI) with respect to overall and severe neurotoxicity. The medications were ranked according to their surface under cumulative ranking curve values. The consistency of direct and indirect evidence was also evaluated. We found that patients with amifostine or vitamin E (VE) treatment exhibited a lower risk of overall neurotoxicity compared to those using the placebo (amifostine: OR = 0.10, 95% CrI: 0.02–0.46; VE: OR = 0.08, 95% CrI: 0.01–0.99). In regard to preventing severe neurotoxicity, glutathione and amifostine treatment appeared to be significantly more effective than the placebo (glutathione: OR = 0.19, 95% CrI: 0.04–0.64; amifostine: OR = 0.12, 95% CrI: 0.02–0.48). In summary, amifostine, VE, and glutathione treatment is considered to be effective in lowering the risk of CICNT. However, further studies which consider safety are required.
Collapse
Affiliation(s)
- Xiying Fu
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Huijie Wu
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinyao Li
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Can Wang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ming Li
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Qianqian Ma
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Yang
- Department of Neurology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
26
|
Starobova H, Vetter I. Pathophysiology of Chemotherapy-Induced Peripheral Neuropathy. Front Mol Neurosci 2017; 10:174. [PMID: 28620280 PMCID: PMC5450696 DOI: 10.3389/fnmol.2017.00174] [Citation(s) in RCA: 376] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/17/2017] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy-induced neuropathy is a common, dose-dependent adverse effect of several antineoplastics. It can lead to detrimental dose reductions and discontinuation of treatment, and severely affects the quality of life of cancer survivors. Clinically, chemotherapy-induced peripheral neuropathy presents as deficits in sensory, motor, and autonomic function which develop in a glove and stocking distribution due to preferential effects on longer axons. The pathophysiological processes are multi-factorial and involve oxidative stress, apoptotic mechanisms, altered calcium homeostasis, axon degeneration and membrane remodeling as well as immune processes and neuroinflammation. This review focusses on the commonly used antineoplastic substances oxaliplatin, cisplatin, vincristine, docetaxel, and paclitaxel which interfere with the cancer cell cycle-leading to cell death and tumor degradation-and cause severe acute and chronic peripheral neuropathies. We discuss drug mechanism of action and pharmacokinetic disposition relevant to the development of peripheral neuropathy, the epidemiology and clinical presentation of chemotherapy-induced neuropathy, emerging insight into genetic susceptibilities as well as current understanding of the pathophysiology and treatment approaches.
Collapse
Affiliation(s)
- Hana Starobova
- Centre for Pain Research, Institute for Molecular Bioscience, University of QueenslandSt Lucia, QLD, Australia
| | - Irina Vetter
- Centre for Pain Research, Institute for Molecular Bioscience, University of QueenslandSt Lucia, QLD, Australia.,School of Pharmacy, University of QueenslandSt Lucia, QLD, Australia
| |
Collapse
|
27
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 2017; 104:144-164. [PMID: 28088622 DOI: 10.1016/j.freeradbiomed.2017.01.004] [Citation(s) in RCA: 626] [Impact Index Per Article: 89.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS), a group of highly reactive ions and molecules, are increasingly being appreciated as powerful signaling molecules involved in the regulation of a variety of biological processes. Indeed, their role is continuously being delineated in a variety of pathophysiological conditions. For instance, cancer cells are shown to have increased ROS levels in comparison to their normal counterparts. This is partly due to an enhanced metabolism and mitochondrial dysfunction in cancer cells. The escalated ROS generation in cancer cells contributes to the biochemical and molecular changes necessary for the tumor initiation, promotion and progression, as well as, tumor resistance to chemotherapy. Therefore, increased ROS in cancer cells may provide a unique opportunity to eliminate cancer cells via elevating ROS to highly toxic levels intracellularly, thereby, activating various ROS-induced cell death pathways, or inhibiting cancer cell resistance to chemotherapy. Such results can be achieved by using agents that either increase ROS generation, or inhibit antioxidant defense, or even a combination of both. In fact, a large variety of anticancer drugs, and some of those currently under clinical trials, effectively kill cancer cells and overcome drug resistance via enhancing ROS generation and/or impeding the antioxidant defense mechanism. This review focuses on our current understanding of the tumor promoting (tumorigenesis, angiogenesis, invasion and metastasis, and chemoresistance) and the tumor suppressive (apoptosis, autophagy, and necroptosis) functions of ROS, and highlights the potential mechanism(s) involved. It also sheds light on a very novel and an actively growing field of ROS-dependent cell death mechanism referred to as ferroptosis.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE; Al Jalila Foundation Research Centre, P.O. Box 300100, Dubai, UAE.
| | - Anees Rahman
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
28
|
Banach M, Juranek JK, Zygulska AL. Chemotherapy-induced neuropathies-a growing problem for patients and health care providers. Brain Behav 2017; 7:e00558. [PMID: 28127506 PMCID: PMC5256170 DOI: 10.1002/brb3.558] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 07/12/2016] [Accepted: 07/18/2016] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Chemotherapy-induced neuropathies are one of the most common side effects of cancer treatment, surpassing bone marrow suppression and kidney dysfunction. Chemotherapy effects on the nervous system vary between different classes of drugs and depend on specific chemical and physical properties of the drug used. The three most neurotoxic classes of anti-cancer drugs are: platinum-based drugs, taxanes, and thalidomide and its analogs; other, less neurotoxic but also commonly used drugs are: bortezomib, ixabepilone, and vinca alkaloids. METHODS Here, in this paper, based on our experience and current knowledge, we provide a short review of the most common, neuropathy-inducing anti-cancer drugs, describe the most prevalent neuropathy symptoms produced by each of them, and outline preventive measures and treatment guidelines for cancer patients suffering from neuropathy and for their health care providers. RESULTS Patients should be encouraged to report any signs of neuropathic pain, alteration in sensory perception, tingling, numbness, burning, increased hot/cold sensitivity and motor dysfunctions as early as possible. If known neurotoxic chemotherapeutics are used, a neurological examination with electrophysiological evaluation should be implemented early in the course of treatment so, both patients and physicians would be better prepared to cope with possible neurotoxic effects. CONCLUSIONS The use of neurotoxic chemotherapeutics should be closely monitored and if clinically permitted, that is, if a patient shows signs of cancer regression, drug doses should be reduced or combined with other less neurotoxic anti-cancer medication. If not counteractive, the use of over the counter antineuropathic supplements such as calcium or magnesium might be encouraged. If physically possible, patients should also be encouraged to exercise regularly and avoid factors that might increase nerve damage such as excessive drinking, smoking, or sitting in a cramped position.
Collapse
Affiliation(s)
- Marta Banach
- Department of Neurology Collegium Medicum Jagiellonian University Krakow Poland
| | - Judyta K Juranek
- Department of Medicine New York University Langone Medical Center New York NY USA; Department of Pathology Faculty of Medical Science University of Warmia and Mazury Olsztyn Poland
| | - Aneta L Zygulska
- Department of Oncology University Hospital Jagiellonian University Krakow Poland
| |
Collapse
|
29
|
Aromolaran KA, Goldstein PA. Ion channels and neuronal hyperexcitability in chemotherapy-induced peripheral neuropathy; cause and effect? Mol Pain 2017; 13:1744806917714693. [PMID: 28580836 PMCID: PMC5480635 DOI: 10.1177/1744806917714693] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 12/18/2022] Open
Abstract
Abstract Cancer is the second leading cause of death worldwide and is a major global health burden. Significant improvements in survival have been achieved, due in part to advances in adjuvant antineoplastic chemotherapy. The most commonly used antineoplastics belong to the taxane, platinum, and vinca alkaloid families. While beneficial, these agents are frequently accompanied by severe side effects, including chemotherapy-induced peripheral neuropathy (CPIN). While CPIN affects both motor and sensory systems, the majority of symptoms are sensory, with pain, tingling, and numbness being the predominant complaints. CPIN not only decreases the quality of life of cancer survivors but also can lead to discontinuation of treatment, thereby adversely affecting survival. Consequently, minimizing the incidence or severity of CPIN is highly desirable, but strategies to prevent and/or treat CIPN have proven elusive. One difficulty in achieving this goal arises from the fact that the molecular and cellular mechanisms that produce CPIN are not fully known; however, one common mechanism appears to be changes in ion channel expression in primary afferent sensory neurons. The processes that underlie chemotherapy-induced changes in ion channel expression and function are poorly understood. Not all antineoplastic agents directly affect ion channel function, suggesting additional pathways may contribute to the development of CPIN Indeed, there are indications that these drugs may mediate their effects through cellular signaling pathways including second messengers and inflammatory cytokines. Here, we focus on ion channelopathies as causal mechanisms for CPIN and review the data from both pre-clinical animal models and from human studies with the aim of facilitating the development of appropriate strategies to prevent and/or treat CPIN.
Collapse
Affiliation(s)
- Kelly A Aromolaran
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
| | - Peter A Goldstein
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
30
|
Moss RW. Should Patients Undergoing Chemotherapy and Radiotherapy Be Prescribed Antioxidants? Integr Cancer Ther 2016; 5:63-82. [PMID: 16484715 DOI: 10.1177/1534735405285882] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
In September 2005, CA: A Cancer Journal for Clinicians published a warning by Gabriella D’Andrea, MD, against the concurrent use of antioxidants with radiotherapy and chemotherapy. However, several deficiencies of the CA article soon became apparent, not least the selective omission of prominent studies that contradicted the author’s conclusions. While acknowledging that only large-scale, randomized trials could provide a valid basis for therapeutic recommendations, the author sometimes relied on laboratory rather than clinical data to support her claim that harm resulted from the concurrent use of antioxidants and chemotherapy. She also sometimes extrapolated from chemoprevention studies rather than those on the concurrent use of antioxidants per se. The article overstated the degree to which the laboratory data diverged in regard to the safety and efficacy of antioxidant therapy: in fact, the preponderance of data suggests a synergistic or at least harmless effect with most high-dose dietary antioxidants and chemotherapy. The practical recommendations made in the article to avoid the general class of antioxidants during chemotherapy are inconsistent, in that if antioxidants were truly a threat to the efficacy of standard therapy, antioxidant-rich foods, especially fruits and vegetables, ought also be proscribed during treatment. Yet no such recommendation is made. Furthermore, the wide-scale use by both medical and radiation oncologists of synthetic antioxidants (eg, amifostine) to control the adverse effects of cytotoxic treatments is similarly overlooked. In sum, this CA article is incomplete: there is far more information available regarding antioxidant supplements as an appropriate adjunctive cancer therapy than is acknowledged. Patients would be well advised to seek the opinion of physicians who are adequately trained and experienced in the intersection of 2 complex fields, that is, chemotherapeutics and nutritional oncology. Physicians whose goal is comprehensive cancer therapy should refer their patients to qualified integrative practitioners who have such training and expertise to guide patients. A blanket rejection of the concurrent use of antioxidants with chemotherapy is not justified by the preponderance of evidence at this time and serves neither the scientific community nor cancer patients.
Collapse
Affiliation(s)
- Ralph W Moss
- Cancer Communications, Lemont, Pennsylvania 16851, USA.
| |
Collapse
|
31
|
Rodgers C, Sanborn C, Taylor O, Gundy P, Pasvogel A, Moore IMK, Hockenberry MJ. Fatigue and Oxidative Stress in Children Undergoing Leukemia Treatment. Biol Res Nurs 2016; 18:515-20. [PMID: 27179013 DOI: 10.1177/1099800416647794] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Fatigue is a frequent and distressing symptom in children undergoing leukemia treatment; however, little is known about factors influencing this symptom. Antioxidants such as glutathione can decrease symptom severity in adult oncology patients, but no study has evaluated antioxidants' effects on symptoms in pediatric oncology patients. This study describes fatigue patterns and associations of fatigue with antioxidants represented by reduced glutathione (GSH) and the reduced/oxidized glutathione (GSH/GSSG) ratio among children receiving leukemia treatment. A repeated measures design assessed fatigue and antioxidants among 38 children from two large U.S. cancer centers. Fatigue was assessed among school-age children and by parent proxy among young children. Antioxidants (GSH and GSH/GSSG ratio) were assessed from cerebrospinal fluid at four phases during leukemia treatment. Young children had a steady decline of fatigue from the end of induction treatment through the continuation phase of treatment, but no significant changes were noted among the school-age children. Mean antioxidant scores varied slightly over time; however, the GSH/GSSG ratios in these children were significantly lower than the normal ratio. Mean GSH/GSSG ratios significantly correlated to fatigue scores of the school-age children during early phases of treatment. Children with low mean GSH/GSSG ratios demonstrated oxidative stress. The low ratios noted early in therapy were significantly correlated with higher fatigue scores during induction and postinduction treatment phases. This finding suggests that increased oxidative stress during the more intensive phases of therapy may explain the experience of fatigue children report.
Collapse
Affiliation(s)
| | - Chelse Sanborn
- Duke Children's Hospital & Health Center, Durham, NC, USA
| | | | - Patricia Gundy
- University of Arizona College of Nursing, Tucson, AZ, USA
| | - Alice Pasvogel
- University of Arizona College of Nursing, Tucson, AZ, USA
| | - Ida M Ki Moore
- University of Arizona College of Nursing, Tucson, AZ, USA
| | | |
Collapse
|
32
|
Research design considerations for chronic pain prevention clinical trials: IMMPACT recommendations. Pain 2016; 156:1184-1197. [PMID: 25887465 DOI: 10.1097/j.pain.0000000000000191] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Although certain risk factors can identify individuals who are most likely to develop chronic pain, few interventions to prevent chronic pain have been identified. To facilitate the identification of preventive interventions, an IMMPACT meeting was convened to discuss research design considerations for clinical trials investigating the prevention of chronic pain. We present general design considerations for prevention trials in populations that are at relatively high risk for developing chronic pain. Specific design considerations included subject identification, timing and duration of treatment, outcomes, timing of assessment, and adjusting for risk factors in the analyses. We provide a detailed examination of 4 models of chronic pain prevention (ie, chronic postsurgical pain, postherpetic neuralgia, chronic low back pain, and painful chemotherapy-induced peripheral neuropathy). The issues discussed can, in many instances, be extrapolated to other chronic pain conditions. These examples were selected because they are representative models of primary and secondary prevention, reflect persistent pain resulting from multiple insults (ie, surgery, viral infection, injury, and toxic or noxious element exposure), and are chronically painful conditions that are treated with a range of interventions. Improvements in the design of chronic pain prevention trials could improve assay sensitivity and thus accelerate the identification of efficacious interventions. Such interventions would have the potential to reduce the prevalence of chronic pain in the population. Additionally, standardization of outcomes in prevention clinical trials will facilitate meta-analyses and systematic reviews and improve detection of preventive strategies emerging from clinical trials.
Collapse
|
33
|
Schloss J, Colosimo M, Vitetta L. New Insights into Potential Prevention and Management Options for Chemotherapy-Induced Peripheral Neuropathy. Asia Pac J Oncol Nurs 2016; 3:73-85. [PMID: 27981142 PMCID: PMC5123533 DOI: 10.4103/2347-5625.170977] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 10/21/2015] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Neurological complications such as chemotherapy-induced peripheral neuropathy (CIPN) and neuropathic pain are frequent side effects of neurotoxic chemotherapy agents. An increasing survival rate and frequent administration of adjuvant chemotherapy treatments involving neurotoxic agents makes it imperative that accurate diagnosis, prevention, and treatment of these neurological complications be implemented. METHODS A consideration was undertaken of the current options regarding protective and treatment interventions for patients undergoing chemotherapy with neurotoxic chemotherapy agent or experience with CIPN. Current knowledge on the mechanism of action has also been identified. The following databases PubMed, the Cochrane Library, Science Direct, Scopus, EMBASE, MEDLINE, CINAHL, CNKI, and Google Scholar were searched for relevant article retrieval. RESULTS A range of pharmaceutical, nutraceutical, and herbal medicine treatments were identified that either showed efficacy or had some evidence of efficacy. Duloxetine was the most effective pharmaceutical agent for the treatment of CIPN. Vitamin E demonstrated potential for the prevention of cisplatin-IPN. Intravenous glutathione for oxaliplatin, Vitamin B6 for both oxaliplatin and cisplatin, and omega 3 fatty acids for paclitaxel have shown protection for CIPN. Acetyl-L-carnitine may provide some relief as a treatment option. Acupuncture may be of benefit for some patients and Gosha-jinki-gan may be of benefit for protection from adverse effects of oxaliplatin induced peripheral neuropathy. CONCLUSIONS Clinicians and researchers acknowledge that there are numerous challenges involved in understanding, preventing, and treating peripheral neuropathy caused by chemotherapeutic agents. New insights into mechanisms of action from chemotherapy agents may facilitate the development of novel preventative and treatment options, thereby enabling medical staff to better support patients by reducing this debilitating side effect.
Collapse
Affiliation(s)
- Janet Schloss
- Mater Private Breast Cancer Centre, Mater Hospital, Brisbane, Australia
- Office of Research, Endeavour College of Natural Health, University of Technology, Brisbane, Australia
| | - Maree Colosimo
- Mater Private Breast Cancer Centre, Mater Hospital, Brisbane, Australia
- Medical Oncology Group of Australia, Clinical Oncology Society of Australia, Queensland Clinical Oncology Group, Brisbane, Australia
| | - Luis Vitetta
- Sydney Medical School, University of Sydney, Sydney 2006, Sydney, Australia
- Medlab Clinical, Sydney, Australia
| |
Collapse
|
34
|
Majithia N, Temkin SM, Ruddy KJ, Beutler AS, Hershman DL, Loprinzi CL. National Cancer Institute-supported chemotherapy-induced peripheral neuropathy trials: outcomes and lessons. Support Care Cancer 2015; 24:1439-47. [PMID: 26686859 DOI: 10.1007/s00520-015-3063-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 12/14/2015] [Indexed: 12/19/2022]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is one of the most common and debilitating complications of cancer treatment. Due to a lack of effective management options for patients with CIPN, the National Cancer Institute (NCI) sponsored a series of trials aimed at both prevention and treatment. A total of 15 such studies were approved, evaluating use of various neuro-modulatory agents which have shown benefit in other neuropathic pain states. Aside from duloxetine, none of the pharmacologic methods demonstrated therapeutic benefit for patients with CIPN. Despite these disappointing results, the series of trials revealed important lessons that have informed subsequent work. Some examples of this include the use of patient-reported symptom metrics, the elimination of traditional--yet unsubstantiated--practice approaches, and the discovery of molecular genetic predictors of neuropathy. Current inquiry is being guided by the results from these large-scale trials, and as such, stands better chance of identifying durable solutions for this treatment-limiting toxicity.
Collapse
Affiliation(s)
- Neil Majithia
- Department of Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Sarah M Temkin
- Community Oncology and Prevention Trials Research Group, Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Kathryn J Ruddy
- Department of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Andreas S Beutler
- Department of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Dawn L Hershman
- Department of Medicine, Department of Epidemiology, Mailman School of Public Health, Columbia University College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, Columbia University, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital, 161 Fort Washington Ave #1068, New York, NY, 10032, USA
| | - Charles L Loprinzi
- Department of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| |
Collapse
|
35
|
Yasin BR, El-Fawal HAN, Mousa SA. Date (Phoenix dactylifera) Polyphenolics and Other Bioactive Compounds: A Traditional Islamic Remedy's Potential in Prevention of Cell Damage, Cancer Therapeutics and Beyond. Int J Mol Sci 2015; 16:30075-90. [PMID: 26694370 PMCID: PMC4691153 DOI: 10.3390/ijms161226210] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 12/20/2022] Open
Abstract
This review analyzes current studies of the therapeutic effects of Phoenix dactylifera, or date palm fruit, on the physiologic system. Specifically, we sought to summarize the effects of its application in preventing cell damage, improving cancer therapeutics and reducing damage caused by conventional chemotherapy. Phoenix dactylifera exhibits potent anti-oxidative properties both in vitro and in vivo. This allows the fruit to prevent depletion of intrinsic protection from oxidative cell damage and assist these defense systems in reducing cell damage. Macroscopically, this mechanism may be relevant to the prevention of various adverse drug events common to chemotherapy including hepatotoxicity, nephrotoxicity, gastrotoxicity, and peripheral neuropathy. While such effects have only been studied in small animal systems, research suggests a potential application to more complex mammalian systems and perhaps a solution to some problems of chemotherapy in hepato-compromised and nephro-compromised patients.
Collapse
Affiliation(s)
- Bibi R Yasin
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| | - Hassan A N El-Fawal
- Neurotoxicology Laboratory, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA.
| |
Collapse
|
36
|
Santabarbara G, Maione P, Rossi A, Gridelli C. Pharmacotherapeutic options for treating adverse effects of Cisplatin chemotherapy. Expert Opin Pharmacother 2015; 17:561-70. [DOI: 10.1517/14656566.2016.1122757] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Ferguson LR, Chen H, Collins AR, Connell M, Damia G, Dasgupta S, Malhotra M, Meeker AK, Amedei A, Amin A, Ashraf SS, Aquilano K, Azmi AS, Bhakta D, Bilsland A, Boosani CS, Chen S, Ciriolo MR, Fujii H, Guha G, Halicka D, Helferich WG, Keith WN, Mohammed SI, Niccolai E, Yang X, Honoki K, Parslow VR, Prakash S, Rezazadeh S, Shackelford RE, Sidransky D, Tran PT, Yang ES, Maxwell CA. Genomic instability in human cancer: Molecular insights and opportunities for therapeutic attack and prevention through diet and nutrition. Semin Cancer Biol 2015; 35 Suppl:S5-S24. [PMID: 25869442 PMCID: PMC4600419 DOI: 10.1016/j.semcancer.2015.03.005] [Citation(s) in RCA: 195] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 03/08/2015] [Accepted: 03/13/2015] [Indexed: 02/06/2023]
Abstract
Genomic instability can initiate cancer, augment progression, and influence the overall prognosis of the affected patient. Genomic instability arises from many different pathways, such as telomere damage, centrosome amplification, epigenetic modifications, and DNA damage from endogenous and exogenous sources, and can be perpetuating, or limiting, through the induction of mutations or aneuploidy, both enabling and catastrophic. Many cancer treatments induce DNA damage to impair cell division on a global scale but it is accepted that personalized treatments, those that are tailored to the particular patient and type of cancer, must also be developed. In this review, we detail the mechanisms from which genomic instability arises and can lead to cancer, as well as treatments and measures that prevent genomic instability or take advantage of the cellular defects caused by genomic instability. In particular, we identify and discuss five priority targets against genomic instability: (1) prevention of DNA damage; (2) enhancement of DNA repair; (3) targeting deficient DNA repair; (4) impairing centrosome clustering; and, (5) inhibition of telomerase activity. Moreover, we highlight vitamin D and B, selenium, carotenoids, PARP inhibitors, resveratrol, and isothiocyanates as priority approaches against genomic instability. The prioritized target sites and approaches were cross validated to identify potential synergistic effects on a number of important areas of cancer biology.
Collapse
Affiliation(s)
| | - Helen Chen
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, Canada
| | - Andrew R Collins
- Department of Nutrition, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Marisa Connell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, Canada
| | - Giovanna Damia
- Department of Oncology, Instituti di Ricovero e Cura a Carattere Scientifico-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Santanu Dasgupta
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, United States
| | | | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Katia Aquilano
- Department of Biology, Università di Roma Tor Vergata, Rome, Italy
| | - Asfar S Azmi
- Department of Biology, University of Rochester, Rochester, United States
| | - Dipita Bhakta
- School of Chemical and BioTechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chandra S Boosani
- Department of BioMedical Sciences, Creighton University, Omaha, NE, United States
| | - Sophie Chen
- Department of Research & Development, Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey, United Kingdom
| | | | - Hiromasa Fujii
- Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Gunjan Guha
- School of Chemical and BioTechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - William G Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Xujuan Yang
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Kanya Honoki
- Department of Orthopaedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | | | - Satya Prakash
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Sarallah Rezazadeh
- Department of Biology, University of Rochester, Rochester, United States
| | - Rodney E Shackelford
- Department of Pathology, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Phuoc T Tran
- Departments of Radiation Oncology & Molecular Radiation Sciences, Oncology and Urology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Christopher A Maxwell
- Department of Pediatrics, University of British Columbia, Michael Cuccione Childhood Cancer Research Program, Child and Family Research Institute, Vancouver, Canada.
| |
Collapse
|
38
|
Śpiewak K, Stochel G, Brindell M. Influence of redox activation of NAMI-A on affinity to serum proteins: transferrin and albumin. J COORD CHEM 2015. [DOI: 10.1080/00958972.2015.1067692] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- K. Śpiewak
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Krakow, Poland
| | - G. Stochel
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Krakow, Poland
| | - M. Brindell
- Faculty of Chemistry, Department of Inorganic Chemistry, Jagiellonian University, Krakow, Poland
| |
Collapse
|
39
|
Avan A, Postma TJ, Ceresa C, Avan A, Cavaletti G, Giovannetti E, Peters GJ. Platinum-induced neurotoxicity and preventive strategies: past, present, and future. Oncologist 2015; 20:411-32. [PMID: 25765877 PMCID: PMC4391771 DOI: 10.1634/theoncologist.2014-0044] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 12/11/2014] [Indexed: 02/07/2023] Open
Abstract
Neurotoxicity is a burdensome side effect of platinum-based chemotherapy that prevents administration of the full efficacious dosage and often leads to treatment withdrawal. Peripheral sensory neurotoxicity varies from paresthesia in fingers to ataxic gait, which might be transient or irreversible. Because the number of patients being treated with these neurotoxic agents is still increasing, the need for understanding the pathogenesis of this dramatic side effect is critical. Platinum derivatives, such as cisplatin and carboplatin, harm mainly peripheral nerves and dorsal root ganglia neurons, possibly because of progressive DNA-adduct accumulation and inhibition of DNA repair pathways (e.g., extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase/stress-activated protein kinase, and p38 mitogen-activated protein kinass), which finally mediate apoptosis. Oxaliplatin, with a completely different pharmacokinetic profile, may also alter calcium-sensitive voltage-gated sodium channel kinetics through a calcium ion immobilization by oxalate residue as a calcium chelator and cause acute neurotoxicity. Polymorphisms in several genes, such as voltage-gated sodium channel genes or genes affecting the activity of pivotal metal transporters (e.g., organic cation transporters, organic cation/carnitine transporters, and some metal transporters, such as the copper transporters, and multidrug resistance-associated proteins), can also influence drug neurotoxicity and treatment response. However, most pharmacogenetics studies need to be elucidated by robust evidence. There are supportive reports about the effectiveness of several neuroprotective agents (e.g., vitamin E, glutathione, amifostine, xaliproden, and venlafaxine), but dose adjustment and/or drug withdrawal seem to be the most frequently used methods in the management of platinum-induced peripheral neurotoxicity. To develop alternative options in the treatment of platinum-induced neuropathy, studies on in vitro models and appropriate trials planning should be integrated into the future design of neuroprotective strategies to find the best patient-oriented solution.
Collapse
Affiliation(s)
- Abolfazl Avan
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tjeerd J Postma
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Cecilia Ceresa
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Guido Cavaletti
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elisa Giovannetti
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Godefridus J Peters
- Departments of Medical Oncology and Neurology, VU University Medical Center, Amsterdam, The Netherlands; Department of Surgery and Translational Medicine, University of Milano-Bicocca, Monza, Italy; Department of New Sciences and Technology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
40
|
Hong G, White J, Zhong L, Carlson LE. Survey of Policies and Guidelines on Antioxidant Use for Cancer Prevention, Treatment, and Survivorship in North American Cancer Centers: What Do Institutions Perceive as Evidence? Integr Cancer Ther 2015; 14:305-17. [PMID: 25716350 DOI: 10.1177/1534735415572884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Health care policies and guidelines that are clear and consistent with research evidence are important for maximizing clinical outcomes. To determine whether cancer centers in Canada and the United States had policies and/or guidelines about antioxidant use, and whether policies were aligned with the evidence base, we reviewed current research evidence in the field, and we undertook a survey of the policies and guidelines on antioxidant use at cancer institutions across North America. METHODS A survey of policies and guidelines on antioxidant use and the development and communication of the policies and guidelines was conducted by contacting cancer institutions in North America. We also conducted a Website search for each institution to explore any online resources. RESULTS Policies and guidelines on antioxidant use were collected from 78 cancer institutions. Few cancer institutions had policies (5%) but most provided guidelines (69%). Antioxidants from diet were generally encouraged at cancer institutions, consistent with the current research evidence. In contrast, specific antioxidant supplements were generally not recommended at cancer institutions. Policies and guidelines were developed using evidence-based methods (53%), by consulting another source (35%), or through discussions/conference (26%), and communicated mainly through online resources (65%) or written handouts (42%). For cancer institutions that had no policy or guideline on antioxidants, lack of information and lack of time were the most frequently cited reasons. CONCLUSIONS Policies and guidelines on antioxidants from diet were largely consistent with the research evidence. Policies and guidelines on antioxidant supplements during treatment were generally more restrictive than the research evidence might suggest, perhaps due to the specificity of results and the inability to generalize findings across antioxidants, adding to the complexity of their optimal and safe use. Improved communication of comprehensive research evidence to cancer institutions may aid in the development of more evidence-based policies and guidelines.
Collapse
Affiliation(s)
- Gyeongyeon Hong
- Tom Baker Cancer Centre-Holy Cross Site, Calgary, Alberta, Canada
| | - Jennifer White
- Tom Baker Cancer Centre-Holy Cross Site, Calgary, Alberta, Canada
| | - Lihong Zhong
- Tom Baker Cancer Centre-Holy Cross Site, Calgary, Alberta, Canada
| | - Linda E Carlson
- Tom Baker Cancer Centre-Holy Cross Site, Calgary, Alberta, Canada University of Calgary Department of Oncology, Cumming School of Medicine, Calgary, Alberta, Canada
| |
Collapse
|
41
|
Fehrenbacher JC. Chemotherapy-Induced Peripheral Neuropathy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:471-508. [DOI: 10.1016/bs.pmbts.2014.12.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
42
|
Friesland A, Weng Z, Duenas M, Massa SM, Longo FM, Lu Q. Amelioration of cisplatin-induced experimental peripheral neuropathy by a small molecule targeting p75 NTR. Neurotoxicology 2014; 45:81-90. [PMID: 25277379 PMCID: PMC4268328 DOI: 10.1016/j.neuro.2014.09.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 08/18/2014] [Accepted: 09/22/2014] [Indexed: 12/18/2022]
Abstract
Cisplatin is an effective and widely used first-line chemotherapeutic drug for treating cancers. However, many patients sustain cisplatin-induced peripheral neuropathy (CIPN), often leading to a reduction in drug dosages or complete cessation of treatment altogether. Therefore, it is important to understand cisplatin mechanisms in peripheral nerve tissue mediating its toxicity and identify signaling pathways for potential intervention. Rho GTPase activation is increased following trauma in several models of neuronal injury. Thus, we investigated whether components of the Rho signaling pathway represent important neuroprotective targets with the potential to ameliorate CIPN and thereby optimize current chemotherapy treatment regimens. We have developed a novel CIPN model in the mouse. Using this model and primary neuronal culture, we determined whether LM11A-31, a small-molecule, orally bioavailable ligand of the p75 neurotrophin receptor (p75(NTR)), can modulate Rho GTPase signaling and reduce CIPN. Von Frey filament analysis of sural nerve function showed that LM11A-31 treatment prevented decreases in peripheral nerve sensation seen with cisplatin treatment. Morphometric analysis of harvested sural nerves revealed that cisplatin-induced abnormal nerve fiber morphology and the decreases in fiber area were alleviated with concurrent LM11A-31 treatment. Cisplatin treatment increased RhoA activity accompanied by the reduced tyrosine phosphorylation of SHP2, which was reversed by LM11A-31. LM11A-31 also countered the effects of calpeptin, which activated RhoA by inhibiting SHP2 tyrosine phosphatase. Therefore, suppression of RhoA signaling by LM11A-31 that modulates p75(NTR) or activates SHP2 tyrosine phosphatase downstream of the NGF receptor enhances neuroprotection in experimental CIPN in mouse model.
Collapse
Affiliation(s)
- Amy Friesland
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Leo Jenkins Cancer Center, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Zhiying Weng
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, China
| | - Maria Duenas
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Stephen M Massa
- Department of Neurology Veterans Administration Medical Center and University of California at San Francisco, San Francisco, CA 94121, USA
| | - Frank M Longo
- Department of Neurology and Neurological Science, Stanford University, Stanford, CA 94305, USA
| | - Qun Lu
- Department of Anatomy and Cell Biology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Leo Jenkins Cancer Center, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
43
|
Zhao J, Gou S, Xu G, Cheng L. Antitumor platinum(II) complexes of N-monoalkyl 1R,2R-diamino-cyclohexanes with 3-(nitrooxy)cyclobutane-1,1-dicarboxylate as a leaving group. Eur J Med Chem 2014; 85:408-17. [DOI: 10.1016/j.ejmech.2014.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 07/31/2014] [Accepted: 08/04/2014] [Indexed: 12/24/2022]
|
44
|
Brief review: Chemotherapy-induced painful peripheral neuropathy (CIPPN): current status and future directions. Can J Anaesth 2014; 61:754-62. [DOI: 10.1007/s12630-014-0171-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/16/2014] [Indexed: 10/25/2022] Open
|
45
|
Hershman DL, Lacchetti C, Dworkin RH, Lavoie Smith EM, Bleeker J, Cavaletti G, Chauhan C, Gavin P, Lavino A, Lustberg MB, Paice J, Schneider B, Smith ML, Smith T, Terstriep S, Wagner-Johnston N, Bak K, Loprinzi CL. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol 2014; 32:1941-67. [PMID: 24733808 DOI: 10.1200/jco.2013.54.0914] [Citation(s) in RCA: 807] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE To provide evidence-based guidance on the optimum prevention and treatment approaches in the management of chemotherapy-induced peripheral neuropathies (CIPN) in adult cancer survivors. METHODS A systematic literature search identified relevant, randomized controlled trials (RCTs) for the treatment of CIPN. Primary outcomes included incidence and severity of neuropathy as measured by neurophysiologic changes, patient-reported outcomes, and quality of life. RESULTS A total of 48 RCTs met eligibility criteria and comprise the evidentiary basis for the recommendations. Trials tended to be small and heterogeneous, many with insufficient sample sizes to detect clinically important differences in outcomes. Primary outcomes varied across the trials, and in most cases, studies were not directly comparable because of different outcomes, measurements, and instruments used at different time points. The strength of the recommendations is based on the quality, amount, and consistency of the evidence and the balance between benefits and harms. RECOMMENDATIONS On the basis of the paucity of high-quality, consistent evidence, there are no agents recommended for the prevention of CIPN. With regard to the treatment of existing CIPN, the best available data support a moderate recommendation for treatment with duloxetine. Although the CIPN trials are inconclusive regarding tricyclic antidepressants (such as nortriptyline), gabapentin, and a compounded topical gel containing baclofen, amitriptyline HCL, and ketamine, these agents may be offered on the basis of data supporting their utility in other neuropathic pain conditions given the limited other CIPN treatment options. Further research on these agents is warranted.
Collapse
Affiliation(s)
- Dawn L Hershman
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Christina Lacchetti
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Robert H Dworkin
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Ellen M Lavoie Smith
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Jonathan Bleeker
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Guido Cavaletti
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Cynthia Chauhan
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Patrick Gavin
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Antoinette Lavino
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Maryam B Lustberg
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Judith Paice
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Bryan Schneider
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Mary Lou Smith
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Tom Smith
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Shelby Terstriep
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Nina Wagner-Johnston
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Kate Bak
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | - Charles L Loprinzi
- Dawn Hershman, Columbia University Medical Center, New York; Robert Dworkin, University of Rochester, Rochester, NY; Christina Lacchetti and Kate Bak, American Society of Clinical Oncology, Alexandria, VA; Ellen M. Lavoie Smith, University of Michigan, Ann Arbor; Patrick Gavin, Marne, MI; Jonathan Bleeker, Sanford University of South Dakota Medical Center, Sioux Falls, SD; Guido Cavaletti, University of Milano-Bicocca, Monza, Italy; Cynthia Chauhan, Wichita, KS; Antoinette Lavino, Massachusetts General North Shore Cancer Center, Danvers, MA; Maryam Lustberg, Ohio State University, Columbus, OH; Judith Paice, Northwestern University, Chicago, IL; Bryan Schneider, Indiana University, Indianapolis, IN; Mary Lou Smith, Research Advocacy Network, Plano, TX; Tom Smith, Johns Hopkins, Baltimore, MD; Shelby Terstriep, Sanford Health, Fargo, ND; Nina Wagner-Johnston, Washington University, St Louis, MO; and Charles Loprinzi, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
46
|
Albers JW, Chaudhry V, Cavaletti G, Donehower RC. Interventions for preventing neuropathy caused by cisplatin and related compounds. THE COCHRANE DATABASE OF SYSTEMATIC REVIEWS 2014. [PMID: 24687190 DOI: 10.1002/14651858.cd005228] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Cisplatin and several related antineoplastic drugs used to treat many types of solid tumours are neurotoxic, and most patients completing a full course of cisplatin chemotherapy develop a clinically detectable sensory neuropathy. Effective neuroprotective therapies have been sought. OBJECTIVES To examine the efficacy and safety of purported chemoprotective agents to prevent or limit the neurotoxicity of cisplatin and related drugs. SEARCH METHODS On 4 March 2013, we searched the Cochrane Neuromuscular Disease Group Specialized Register, CENTRAL, MEDLINE, EMBASE, LILACS, and CINAHL Plus for randomised trials designed to evaluate neuroprotective agents used to prevent or limit neurotoxicity of cisplatin and related drugs among human patients. SELECTION CRITERIA We included randomised controlled trials (RCTs) or quasi-RCTs in which the participants received chemotherapy with cisplatin or related compounds, with a potential chemoprotectant (acetylcysteine, amifostine, adrenocorticotrophic hormone (ACTH), BNP7787, calcium and magnesium (Ca/Mg), diethyldithiocarbamate (DDTC), glutathione, Org 2766, oxcarbazepine, or vitamin E) compared to placebo, no treatment, or other treatments. We considered trials in which participants underwent evaluation zero to six months after completing chemotherapy using quantitative sensory testing (the primary outcome) or other measures including nerve conduction studies or neurological impairment rating using validated scales (secondary outcomes). DATA COLLECTION AND ANALYSIS Two review authors assessed each study, extracted the data and reached consensus, according to standard Cochrane methodology. MAIN RESULTS As of 2013, the review includes 29 studies describing nine possible chemoprotective agents, as well as description of two published meta-analyses. Among these trials, there were sufficient data in some instances to combine the results from different studies, most often using data from secondary non-quantitative measures. Nine of the studies were newly included at this update. Few of the included studies were at a high risk of bias overall, although often there was too little information to make an assessment. At least two review authors performed a formal review of an additional 44 articles but we did not include them in the final review for a variety of reasons.Of seven eligible amifostine trials (743 participants in total), one used quantitative sensory testing (vibration perception threshold) and demonstrated a favourable outcome in terms of amifostine neuroprotection, but the vibration perception threshold result was based on data from only 14 participants receiving amifostine who completed the post-treatment evaluation and should be regarded with caution. Furthermore the change measured was subclinical. None of the three eligible Ca/Mg trials (or four trials if a single retrospective study was included) described our primary outcome measures. The four Ca/Mg trials included a total of 886 participants. Of the seven eligible glutathione trials (387 participants), one used quantitative sensory testing but reported only qualitative analyses. Four eligible Org 2766 trials (311 participants) employed quantitative sensory testing but reported disparate results; meta-analyses of three of these trials using comparable measures showed no significant vibration perception threshold neuroprotection. The remaining trial reported only descriptive analyses. Similarly, none of the three eligible vitamin E trials (246 participants) reported quantitative sensory testing. The eligible single trials involving acetylcysteine (14 participants), diethyldithiocarbamate (195 participants), oxcarbazepine (32 participants), and retinoic acid (92 participants) did not perform quantitative sensory testing. In all, this review includes data from 2906 participants. However, only seven trials reported data for the primary outcome measure of this review, (quantitative sensory testing) and only nine trials reported our objective secondary measure, nerve conduction test results. Additionally, methodological heterogeneity precluded pooling of the results in most cases. Nonetheless, a larger number of trials reported the results of secondary (non-quantitative and subjective) measures such as the National Cancer Institute Common Toxicity Criteria (NCI-CTC) for neuropathy (15 trials), and these results we pooled and reported as meta-analysis. Amifostine showed a significantly reduced risk of developing neurotoxicity NCI-CTC (or equivalent) ≥ 2 compared to placebo (RR 0.26, 95% CI 0.11 to 0.61). Glutathione was also efficacious with an RR of 0.29 (95% CI 0.10 to 0.85). In three vitamin E studies subjective measures not suitable for combination in meta analysis each favoured vitamin E. For other interventions the qualitative toxicity measures were either negative (N-acetyl cysteine, Ca/Mg, DDTC and retinoic acid) or not evaluated (oxcarbazepine and Org 2766).Adverse events were infrequent or not reported for most interventions. Amifostine was associated with transient hypotension in 8% to 62% of participants, retinoic acid with hypocalcaemia in 11%, and approximately 20% of participantss withdrew from treatment with DDTC because of toxicity. AUTHORS' CONCLUSIONS At present, the data are insufficient to conclude that any of the purported chemoprotective agents (acetylcysteine, amifostine, calcium and magnesium, diethyldithiocarbamate, glutathione, Org 2766, oxcarbazepine, retinoic acid, or vitamin E) prevent or limit the neurotoxicity of platin drugs among human patients, as determined using quantitative, objective measures of neuropathy. Amifostine, calcium and magnesium, glutathione, and vitamin E showed modest but promising (borderline statistically significant) results favouring their ability to reduce the neurotoxicity of cisplatin and related chemotherapies, as measured using secondary, non-quantitative and subjective measures such as the NCI-CTC neuropathy grading scale. Among these interventions, the efficacy of only vitamin E was evaluated using quantitative nerve conduction studies; the results were negative and did not support the positive findings based on the qualitative measures. In summary, the present studies are limited by the small number of participants receiving any particular agent, a lack of objective measures of neuropathy, and differing results among similar trials, which make it impossible to conclude that any of the neuroprotective agents tested prevent or limit the neurotoxicity of platinum drugs.
Collapse
Affiliation(s)
- James W Albers
- Department of Neurology, University of Michigan, 1C325/0032 University Hospital, 1500 E. Medical Center Drive, Box 0316, Ann Arbor, USA, MI 48109-0032
| | | | | | | |
Collapse
|
47
|
Albers JW, Chaudhry V, Cavaletti G, Donehower RC. Interventions for preventing neuropathy caused by cisplatin and related compounds. Cochrane Database Syst Rev 2014; 2014:CD005228. [PMID: 24687190 PMCID: PMC10891440 DOI: 10.1002/14651858.cd005228.pub4] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Cisplatin and several related antineoplastic drugs used to treat many types of solid tumours are neurotoxic, and most patients completing a full course of cisplatin chemotherapy develop a clinically detectable sensory neuropathy. Effective neuroprotective therapies have been sought. OBJECTIVES To examine the efficacy and safety of purported chemoprotective agents to prevent or limit the neurotoxicity of cisplatin and related drugs. SEARCH METHODS On 4 March 2013, we searched the Cochrane Neuromuscular Disease Group Specialized Register, CENTRAL, MEDLINE, EMBASE, LILACS, and CINAHL Plus for randomised trials designed to evaluate neuroprotective agents used to prevent or limit neurotoxicity of cisplatin and related drugs among human patients. SELECTION CRITERIA We included randomised controlled trials (RCTs) or quasi-RCTs in which the participants received chemotherapy with cisplatin or related compounds, with a potential chemoprotectant (acetylcysteine, amifostine, adrenocorticotrophic hormone (ACTH), BNP7787, calcium and magnesium (Ca/Mg), diethyldithiocarbamate (DDTC), glutathione, Org 2766, oxcarbazepine, or vitamin E) compared to placebo, no treatment, or other treatments. We considered trials in which participants underwent evaluation zero to six months after completing chemotherapy using quantitative sensory testing (the primary outcome) or other measures including nerve conduction studies or neurological impairment rating using validated scales (secondary outcomes). DATA COLLECTION AND ANALYSIS Two review authors assessed each study, extracted the data and reached consensus, according to standard Cochrane methodology. MAIN RESULTS As of 2013, the review includes 29 studies describing nine possible chemoprotective agents, as well as description of two published meta-analyses. Among these trials, there were sufficient data in some instances to combine the results from different studies, most often using data from secondary non-quantitative measures. Nine of the studies were newly included at this update. Few of the included studies were at a high risk of bias overall, although often there was too little information to make an assessment. At least two review authors performed a formal review of an additional 44 articles but we did not include them in the final review for a variety of reasons.Of seven eligible amifostine trials (743 participants in total), one used quantitative sensory testing (vibration perception threshold) and demonstrated a favourable outcome in terms of amifostine neuroprotection, but the vibration perception threshold result was based on data from only 14 participants receiving amifostine who completed the post-treatment evaluation and should be regarded with caution. Furthermore the change measured was subclinical. None of the three eligible Ca/Mg trials (or four trials if a single retrospective study was included) described our primary outcome measures. The four Ca/Mg trials included a total of 886 participants. Of the seven eligible glutathione trials (387 participants), one used quantitative sensory testing but reported only qualitative analyses. Four eligible Org 2766 trials (311 participants) employed quantitative sensory testing but reported disparate results; meta-analyses of three of these trials using comparable measures showed no significant vibration perception threshold neuroprotection. The remaining trial reported only descriptive analyses. Similarly, none of the three eligible vitamin E trials (246 participants) reported quantitative sensory testing. The eligible single trials involving acetylcysteine (14 participants), diethyldithiocarbamate (195 participants), oxcarbazepine (32 participants), and retinoic acid (92 participants) did not perform quantitative sensory testing. In all, this review includes data from 2906 participants. However, only seven trials reported data for the primary outcome measure of this review, (quantitative sensory testing) and only nine trials reported our objective secondary measure, nerve conduction test results. Additionally, methodological heterogeneity precluded pooling of the results in most cases. Nonetheless, a larger number of trials reported the results of secondary (non-quantitative and subjective) measures such as the National Cancer Institute Common Toxicity Criteria (NCI-CTC) for neuropathy (15 trials), and these results we pooled and reported as meta-analysis. Amifostine showed a significantly reduced risk of developing neurotoxicity NCI-CTC (or equivalent) ≥ 2 compared to placebo (RR 0.26, 95% CI 0.11 to 0.61). Glutathione was also efficacious with an RR of 0.29 (95% CI 0.10 to 0.85). In three vitamin E studies subjective measures not suitable for combination in meta analysis each favoured vitamin E. For other interventions the qualitative toxicity measures were either negative (N-acetyl cysteine, Ca/Mg, DDTC and retinoic acid) or not evaluated (oxcarbazepine and Org 2766).Adverse events were infrequent or not reported for most interventions. Amifostine was associated with transient hypotension in 8% to 62% of participants, retinoic acid with hypocalcaemia in 11%, and approximately 20% of participantss withdrew from treatment with DDTC because of toxicity. AUTHORS' CONCLUSIONS At present, the data are insufficient to conclude that any of the purported chemoprotective agents (acetylcysteine, amifostine, calcium and magnesium, diethyldithiocarbamate, glutathione, Org 2766, oxcarbazepine, retinoic acid, or vitamin E) prevent or limit the neurotoxicity of platin drugs among human patients, as determined using quantitative, objective measures of neuropathy. Amifostine, calcium and magnesium, glutathione, and vitamin E showed modest but promising (borderline statistically significant) results favouring their ability to reduce the neurotoxicity of cisplatin and related chemotherapies, as measured using secondary, non-quantitative and subjective measures such as the NCI-CTC neuropathy grading scale. Among these interventions, the efficacy of only vitamin E was evaluated using quantitative nerve conduction studies; the results were negative and did not support the positive findings based on the qualitative measures. In summary, the present studies are limited by the small number of participants receiving any particular agent, a lack of objective measures of neuropathy, and differing results among similar trials, which make it impossible to conclude that any of the neuroprotective agents tested prevent or limit the neurotoxicity of platinum drugs.
Collapse
Affiliation(s)
- James W Albers
- Department of Neurology, University of Michigan, 1C325/0032 University Hospital, 1500 E. Medical Center Drive, Box 0316, Ann Arbor, USA, MI 48109-0032
| | | | | | | |
Collapse
|
48
|
Leal AD, Qin R, Atherton PJ, Haluska P, Behrens RJ, Tiber CH, Watanaboonyakhet P, Weiss M, Adams PT, Dockter TJ, Loprinzi CL. North Central Cancer Treatment Group/Alliance trial N08CA-the use of glutathione for prevention of paclitaxel/carboplatin-induced peripheral neuropathy: a phase 3 randomized, double-blind, placebo-controlled study. Cancer 2014; 120:1890-7. [PMID: 24619793 DOI: 10.1002/cncr.28654] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/04/2013] [Accepted: 12/20/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a significant side effect of taxane and platinum-based chemotherapy. Several studies have supported the potential benefit of glutathione for the prevention of platinum-induced CIPN. The current trial was designed to determine whether glutathione would prevent CIPN as a result of carboplatin/paclitaxel therapy. METHODS In total, 185 patients who received treatment with paclitaxel and carboplatin were accrued between December 4, 2009 and December 19, 2011. Patients were randomized to receive either placebo (n = 91) or 1.5 g/m(2) glutathione (n = 94) over 15 minutes immediately before chemotherapy. CIPN was assessed using the European Organization for Research and Treatment of Cancer Quality-of-Life (EORTC-QLQ) 20-item, CIPN-specific (CIPN20) sensory subscale and the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), version 4.0. RESULTS There were no statistically significant differences between the 2 study arms with regard to: 1) peripheral neurotoxicity, as assessed using both the EORTC-QLQ-CIPN20 (P = .21) and the CTCAE scales (P = .449 for grade ≥2 neurotoxicity; P = .039 for time to development of grade ≥2 neuropathy, in favor of the placebo); 2) the degree of paclitaxel acute pain syndrome (P = .30 for patients who received paclitaxel every 3-4 weeks and P = .002, in favor of the placebo, for patients who received weekly paclitaxel); 3) the time to disease progression (P = .63); or 4) apparent toxicities. Subgroup analyses did not reveal any evidence of benefit in any particular subgroup. CONCLUSIONS The results from this study do not support the use of glutathione for the prevention of paclitaxel/carboplatin-induced CIPN.
Collapse
Affiliation(s)
- Alexis D Leal
- Department of Internal Medicine, Mayo Clinic Rochester, Rochester, Minnesota
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
A phase III randomized, placebo-controlled study of topical amitriptyline and ketamine for chemotherapy-induced peripheral neuropathy (CIPN): a University of Rochester CCOP study of 462 cancer survivors. Support Care Cancer 2014; 22:1807-14. [PMID: 24531792 DOI: 10.1007/s00520-014-2158-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 01/30/2014] [Indexed: 02/04/2023]
Abstract
PURPOSE Chemotherapy-induced peripheral neuropathy (CIPN) occurs in as high as 70% of patients receiving certain types of chemotherapy agents. The FDA has yet to approve a therapy for CIPN. The aim of this multicenter, phase III, randomized, double-blind, placebo-controlled trial was to investigate the efficacy of 2% ketamine plus 4% amitriptyline (KA) cream for reducing CIPN. METHODS Cancer survivors who completed chemotherapy at least 1 month prior and had CIPN (>4 out of 10) were enrolled (N=462). CIPN was assessed using average scores from a 7-day daily diary that asks patients to rate the average "pain, numbness, or tingling in [their] hands and feet over the past 24 h" on an 11-point numeric rating scale at baseline and 6 weeks post intervention. ANCOVA was used to measure differences in 6-week CIPN with effects including baseline CIPN, KA treatment arm, and previous taxane therapy (Y/N). RESULTS The KA treatment showed no effect on 6-week CIPN scores (adjusted mean difference=-0.17, p=0.363). CONCLUSIONS This study suggests that KA cream does not decrease CIPN symptoms in cancer survivors.
Collapse
|
50
|
Areti A, Yerra VG, Naidu V, Kumar A. Oxidative stress and nerve damage: role in chemotherapy induced peripheral neuropathy. Redox Biol 2014; 2:289-95. [PMID: 24494204 PMCID: PMC3909836 DOI: 10.1016/j.redox.2014.01.006] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 01/08/2014] [Accepted: 01/09/2014] [Indexed: 12/17/2022] Open
Abstract
Peripheral neuropathy is a severe dose limiting toxicity associated with cancer chemotherapy. Ever since it was identified, the clear pathological mechanisms underlying chemotherapy induced peripheral neuropathy (CIPN) remain sparse and considerable involvement of oxidative stress and neuroinflammation has been realized recently. Despite the empirical use of antioxidants in the therapy of CIPN, the oxidative stress mediated neuronal damage in peripheral neuropathy is still debatable. The current review focuses on nerve damage due to oxidative stress and mitochondrial dysfunction as key pathogenic mechanisms involved in CIPN. Oxidative stress as a central mediator of apoptosis, neuroinflammation, metabolic disturbances and bioenergetic failure in neurons has been highlighted in this review along with a summary of research on dietary antioxidants and other nutraceuticals which have undergone prospective controlled clinical trials in patients undergoing chemotherapy. Oxidative stress contributes to the pathophysiology of chemotherapy induced peripheral neuropathies (CIPN). Mitotoxicity and mitochondrial dysfunction contribute to amplified oxidative stress. Pharmacological interventions targeted at maintenance of mitochondrial health and function may be beneficial against CIPN.
Collapse
Affiliation(s)
- Aparna Areti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Hyderabad (NIPER-H), Bala Nagar, Hyderabad, AP 500037, India
| | - Veera Ganesh Yerra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Hyderabad (NIPER-H), Bala Nagar, Hyderabad, AP 500037, India
| | - Vgm Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Hyderabad (NIPER-H), Bala Nagar, Hyderabad, AP 500037, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Hyderabad (NIPER-H), Bala Nagar, Hyderabad, AP 500037, India
| |
Collapse
|