Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Transl Oncol ; 42: 101867, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38308919

ABSTRACT

The current study compared the levels and possible associations between systemic soluble immune checkpoints (sICPs, n = 17) and a group of humoral modulators of immune suppressor cells (n = 7) in a cohort of patients with basal cell carcinoma (BCC, n = 40) and a group of healthy control subjects (n = 20). The seven humoral modulators of immunosuppressor cells were represented by the enzymes, arginase 1 and fibroblast activation protein (FAP), the chemokine, RANTES (CCL5) and the cytokines, interleukin-10 and transforming growth factor-ß1 (TGF-ß1), as well as the M2-type macrophage markers, soluble CD163 (sCD163) and sCD206. The plasma levels of six co-inhibitory sICPs, sCTLA-4, sLAG-3, sPD-1, sPD-L1, sTIM-3 and sPD-L2 were significantly elevated in the cohort of BCC patients (p<0.001-p<0.00001), while that of sBTLA was significantly decreased (p<0.006). Of the co-stimulatory sICPs, sCD27 and sGITR were significantly increased (p<0.0002 and p<0.0538) in the cohort of BCC patients, while the others were essentially comparable with those of the control participants; of the dual active sICPs, sHVEM was significantly elevated (p<0.00001) and TLR2 comparable with the control group. A correlation heat map revealed selective, strong associations of TGF-ß1 with seven co-stimulatory (z = 0.618468-0.768131) and four co-inhibitory (z = 0.674040-0.808365) sICPs, as well as with sTLR2 (z = 0.696431). Notwithstanding the association of BCC with selective elevations in the levels of a large group of co-inhibitory sICPs, our novel findings also imply the probable involvement of TGF-ß1 in driving immunosuppression in this malignancy, possibly via activation of regulatory T cells. Notably, these abnormalities were present in patients with either newly diagnosed or recurrent disease.

2.
Support Care Cancer ; 32(1): 36, 2023 Dec 18.
Article in English | MEDLINE | ID: mdl-38105286

ABSTRACT

PURPOSE: This review is an update of the MASCC/ESMO 2015 recommendations for the prophylaxis of acute and delayed nausea and vomiting induced by multiple-day chemotherapy, high-dose chemotherapy, and breakthrough nausea and vomiting. METHODS: A systematic literature search was conducted using PubMed from June 1, 2015, through February 1, 2023. RESULTS: We identified 56 references (16 were duplications or invalid), leaving 40 manuscripts for this search. The panel classified level I evidence (three manuscripts) and level II evidence (14 manuscripts). High-dose chemotherapy and stem cell transplant were discussed in four of these manuscripts, and multiple-day chemotherapy treatment in 15. Some manuscripts covered both topics. Additionally, a search for breakthrough nausea and vomiting resulted in 12 "hits." No new relevant studies were identified. CONCLUSIONS: The recommendations for patients receiving high-dose chemotherapy with stem cell transplants and patients undergoing multiple-day cisplatin were updated. For patients receiving high-dose chemotherapy for stem cell transplant, a combination of a 5-HT3 receptor antagonist with dexamethasone and aprepitant is recommended. Olanzapine could be considered part of the antiemetic regimen. Patients receiving multiple-day cisplatin should receive a 5-HT3 receptor antagonist plus dexamethasone plus aprepitant plus olanzapine. For patients experiencing breakthrough nausea and vomiting, the available evidence suggests using a single dose of olanzapine daily for 3 days.


Subject(s)
Antiemetics , Antineoplastic Agents , Humans , Aprepitant/therapeutic use , Olanzapine/therapeutic use , Cisplatin/adverse effects , Consensus , Serotonin/adverse effects , Antineoplastic Agents/therapeutic use , Vomiting/chemically induced , Vomiting/prevention & control , Vomiting/drug therapy , Nausea/chemically induced , Nausea/prevention & control , Nausea/drug therapy , Antiemetics/therapeutic use , Dexamethasone/therapeutic use
3.
Support Care Cancer ; 31(12): 628, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37828258

ABSTRACT

PURPOSE: Limited knowledge is available on the incidence of febrile neutropenia (FN) in intermediate-risk patients and the rationale for use of granulocyte colony-stimulating factor (G-CSF) in these patients. We aimed to estimate the rate at which patients associated with intermediate risk (10-20%) of FN would develop ≥ 1 episode of FN with a commonly used chemotherapy regimen in clinical practice. METHODS: This prospective, real-world, observational, multinational, multicenter study (December 2016-October 2019) recruited patients with solid tumors or Hodgkin's/non-Hodgkin's lymphoma. Patients receiving chemotherapy with intermediate risk of FN, but not G-CSF as primary prophylaxis were included and observed for the duration of the chemotherapy (≤ 6 cycles and ≤ 30 days after the last chemotherapy administration). RESULTS: In total, 364 patients (median age, 56 years) with 1601 cycles of chemotherapy were included in the analysis. The incidence of FN was 5% in cycle 1, 3% in cycles 2-3, and 1% in cycles 4-6. The rate of patients with ≥ 1 episode of FN was 9%, and 59% of FN events were reported during cycle 1. The rate of grade 4 neutropenia in cycle 1 was 11%, and 15% of patients experienced ≥ 1 episode of grade 4 neutropenia. CONCLUSIONS: Overall, the incidence of FN was low, with a high incidence in cycle 1 and a decrease in the subsequent cycles. These results provide the real FN risk for common chemotherapy regimens in patients generally excluded from clinical trials. Prophylactic G-CSF in intermediate-risk patients could be considered as per clinician's judgement.


Subject(s)
Febrile Neutropenia , Neoplasms , Humans , Middle Aged , Prospective Studies , Neoplasms/drug therapy , Neoplasms/etiology , Granulocyte Colony-Stimulating Factor/therapeutic use , Medical Oncology , Febrile Neutropenia/chemically induced , Febrile Neutropenia/epidemiology , Febrile Neutropenia/prevention & control , Antineoplastic Combined Chemotherapy Protocols/adverse effects
4.
Front Oncol ; 13: 1097309, 2023.
Article in English | MEDLINE | ID: mdl-37064132

ABSTRACT

Neoadjuvant chemotherapy (NAC) may alter the immune landscape of patients with early breast cancer (BC), potentially setting the scene for more effective implementation of checkpoint-targeted immunotherapy. This issue has been investigated in the current study in which alterations in the plasma concentrations of 16 soluble co-stimulatory and co-inhibitory, immune checkpoints were measured sequentially in a cohort of newly diagnosed, early BC patients (n=72), pre-treatment, post-NAC and post-surgery using a Multiplex® bead array platform. Relative to a group of healthy control subjects (n=45), the median pre-treatment levels of five co-stimulatory (CD27, CD40, GITRL, ICOS, GITR) and three co-inhibitory (TIM-3, CTLA-4, PD-L1) soluble checkpoints were significantly lower in the BC patients vs. controls (p<0.021-p<0.0001; and p<0.008-p<0.00001, respectively). Following NAC, the plasma levels of six soluble co-stimulatory checkpoints (CD28, CD40, ICOS, CD27, CD80, GITR), all involved in activation of CD8+ cytotoxic T cells, were significantly increased (p<0.04-p<0.00001), comparable with control values and remained at these levels post-surgery. Of the soluble co-inhibitory checkpoints, three (LAG-3, PD-L1, TIM-3) increased significantly post-NAC, reaching levels significantly greater than those of the control group. PD-1 remained unchanged, while BTLA and CTLA-4 decreased significantly (p<0.03 and p<0.00001, respectively). Normalization of soluble co-stimulatory immune checkpoints is seemingly indicative of reversal of systemic immune dysregulation following administration of NAC in early BC, while recovery of immune homeostasis may explain the increased levels of several negative checkpoint proteins, albeit with the exceptions of CTLA-4 and PD-1. Although a pathological complete response (pCR) was documented in 61% of patients (mostly triple-negative BC), surprisingly, none of the soluble immune checkpoints correlated with the pCR, either pre-treatment or post-NAC. Nevertheless, in the case of the co-stimulatory ICMs, these novel findings are indicative of the immune-restorative potential of NAC in early BC, while in the case of the co-inhibitory ICMs, elevated levels of soluble PD-L1, LAG-3 and TIM-3 post-NAC underscore the augmentative immunotherapeutic promise of targeting these molecules, either individually or in combination, as a strategy, which may contribute to the improved management of early BC.

5.
Front Immunol ; 13: 823842, 2022.
Article in English | MEDLINE | ID: mdl-35677046

ABSTRACT

Breast cancer cells exploit the up-regulation or down-regulation of immune checkpoint proteins to evade anti-tumor immune responses. To explore the possible involvement of this mechanism in promoting systemic immunosuppression, the pre-treatment levels of soluble co-inhibitory and co-stimulatory immune checkpoint molecules, as well as those of cytokines, chemokines, and growth factors were measured in 98 newly diagnosed breast cancer patients and compared with those of 45 healthy controls using multiplex bead array and ELISA technologies. Plasma concentrations of the co-stimulatory immune checkpoints, GITR, GITRL, CD27, CD28, CD40, CD80, CD86 and ICOS, as well as the co-inhibitory molecules, PD-L1, CTLA-4 and TIM-3, were all significantly lower in early breast cancer patients compared to healthy controls, as were those of HVEM and sTLR-2, whereas the plasma concentrations of CX3CL1 (fractalkine), CCL5 (RANTES) and those of the growth factors, M-CSF, FGF-21 and GDF-15 were significantly increased. However, when analyzed according to the patients' breast cancer characteristics, these being triple negative breast cancer (TNBC) vs. non-TNBC, tumor size, stage, nodal status and age, no significant differences were detected between the plasma levels of the various immune checkpoint molecules, cytokines, chemokines and growth factors. Additionally, none of these biomarkers correlated with pathological complete response. This study has identified low plasma levels of soluble co-stimulatory and co-inhibitory immune checkpoint molecules in newly diagnosed, non-metastatic breast cancer patients compared to healthy controls, which is a novel finding seemingly consistent with a state of systemic immune dysregulation. Plausible mechanisms include an association with elevated levels of M-CSF and CCL5, implicating the involvement of immune suppressor cells of the M2-macrophage/monocyte phenotype as possible drivers of this state of systemic immune quiescence/dysregulation.


Subject(s)
Breast Neoplasms , Immune Checkpoint Proteins , Breast Neoplasms/immunology , Breast Neoplasms/physiopathology , Chemokine CCL5/blood , Female , Humans , Immune Checkpoint Proteins/blood , Macrophage Colony-Stimulating Factor/blood
6.
Cancers (Basel) ; 14(10)2022 May 20.
Article in English | MEDLINE | ID: mdl-35626126

ABSTRACT

BACKGROUND: Tumor-infiltrating lymphocytes are associated with a better prognosis in early triple-negative breast cancer (TNBC). These cells can be enumerated in situ by the "Immunoscore Clinical Research" (ISCR). The original Immunoscore® is a prognostic tool that categorizes the densities of CD3+ and CD8+ cells in both the invasive margin (IM) and center of the tumor (CT) in localized colon cancer, yielding a five-tiered classification (0-4). We evaluated the prognostic potential of ISCR and pathological complete response (pCR) following neoadjuvant chemotherapy (NACT). METHODS: The cohort included 53 TNBC, 32 luminal BC, and 18 HER2-positive BC patients undergoing NACT. Pre-treatment tumor biopsies were immune-stained for CD3+ and CD8+ T-cell markers. Quantitative analysis of these cells in different tumor locations was performed using computer-assisted image analysis. RESULTS: The pCR rate was 44%. Univariate analysis showed that primary tumor size, estrogen-receptor negative, progesterone-receptor negative, luminal vs. HER2-positive vs. TNBC, high Ki-67, high densities (cells/mm2) of CD3 CT, CD8+ CT, CD3+ IM, and CD8+ IM cells were associated with a high pCR. ISCR was associated with pCR following NACT. A multivariate model consisting of ISCR and the significant variables from the univariate analysis showed a significant trend for ISCR; however, the low sample size did not provide enough power for the model to be included in this study. CONCLUSIONS: These results revealed a significant prognostic role for the spatial distributions of the CD3+, and CD8+ lymphocytes, as well as the ISCR in relation to pCR following NACT.

7.
Transl Oncol ; 19: 101384, 2022 May.
Article in English | MEDLINE | ID: mdl-35255355

ABSTRACT

Although co-inhibitory immune checkpoint proteins are primarily involved in promoting cell-cell interactions that suppress adaptive immunity, especially tumor immunity, the soluble cell-free variants of these molecules are also detectable in the circulation of cancer patients where they retain immunosuppressive activity. Nevertheless, little is known about the systemic levels of these soluble co-inhibitory immune checkpoints in patients with various subtypes of basal cell carcinoma (BCC), which is the most invasive and treatment-resistant type of this most commonly-occurring malignancy. In the current study, we have measured the systemic concentrations of five prominent co-inhibitory immune checkpoints, namely CTLA-4, LAG-3, PD-1/PD-L1 and TIM-3, as well as those of C-reactive protein (CRP) and vitamin D (VD), in a cohort of patients (n = 40) with BCC, relative to those of a group of control participants, using the combination of multiplex bead array, laser nephelometry and ELISA technologies, respectively. The median systemic concentrations of CRP and VD were comparable between the two groups; however, those of all five immune checkpoints were significantly elevated (P = 0.0184 - P = < 0.00001), with those of CTLA-4 and PD-1 being highly correlated (r = 0.87; P < 0.00001). This seemingly novel finding not only identifies the existence of significant systemic immunosuppression in BCC, but also underscores the therapeutic promise of immune checkpoint targeted therapy, as well as the potential of these proteins to serve as prognostic/predictive biomarkers in BCC.

9.
Ann Palliat Med ; 10(3): 2679-2686, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33549001

ABSTRACT

BACKGROUND: The magnitude of the incidence and impact of nausea on patients receiving intravenous chemotherapy seems to be underestimated by healthcare professionals. Development of effective anti-emetic treatment has contributed to the resolution of chemotherapy-induced nausea and vomiting (CINV). However, there is a concern that vomiting has been the initial focus of anti-emetic research and nausea was perceived as a secondary endpoint. Through focusing on the incidence of nausea independently of the incidence of vomiting, valuable information has been gained on this distressing side effect, including identifying patient risk factors contributing to the increased experience of nausea. METHODS: The study followed a prospective, observational study design in a private oncology centre in Johannesburg, South-Africa. Ethical approval was obtained before commencement of the study, followed by the recruitment of one hundred patients over a seven-month period. Patient-reported outcome measures (PROMs) were used to measure nausea with an amended version of the Multinational Association of Supportive Care in Cancer antiemesis tool (MAT). Patients documented information in their diaries on the incidence, duration and severity of nausea, during the acute phase (0-24 hours), the delayed phase (25-120 hours), day 7 and day 10 after infusion of chemotherapy, with episodes of vomiting being recorded as a secondary endpoint. The demographic and clinical variables of the subjects, as well as patient risk factors known to cause CINV, were tabulated and summarised using descriptive statistics. RESULTS: The population consisted of 68 females and 27 males with a mean age of 57 years (25-84 years). The emetogenicity of chemotherapy regimens administered were well represented with 26.3% low emetogenic chemotherapy, 25.3% moderately emetogenic chemotherapy and 48.4% highly emetogenic chemotherapy (HEC). Despite all patients receiving guideline consistent CINV prophylaxis, nausea was still experienced by 57.9% patients during cycle one, 50.6% patients during cycle two and 45.6% patients during cycle three. The incidence of patients experiencing nausea (in the absence of vomiting) was 35%, compared to 2% of patients experiencing vomiting (in the absence of nausea). Patient characteristics with a known risk to impact CINV were documented, and significant impact in this study was found in female gender, age <60 years, history of motion sickness and history of morning sickness. CONCLUSIONS: Guideline consistent CINV prophylaxes seem to have vomiting under control for most patients receiving intravenous chemotherapy. Nausea, however, still seems to be a persistent adverse event during treatment. Female gender, age <60 years, history of motion sickness and history of morning sickness increases the risk of experiencing nausea. A different approach is needed to manage nausea in the clinic setting, along with standardised tools to measure nausea specifically. More studies need to be done with nausea as the primary endpoint to address this ongoing medical need.


Subject(s)
Antiemetics , Antineoplastic Agents , Neoplasms , Antiemetics/therapeutic use , Antineoplastic Agents/adverse effects , Female , Humans , Incidence , Male , Middle Aged , Nausea/chemically induced , Nausea/drug therapy , Nausea/epidemiology , Neoplasms/drug therapy , Prospective Studies , South Africa , Vomiting/chemically induced , Vomiting/drug therapy , Vomiting/epidemiology
10.
Lung Cancer Manag ; 9(3): LMT37, 2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32774468

ABSTRACT

AIM: We investigated the prognostic potential of pretherapy measurement of the neutrophil/lymphocyte ratio (NLR) in patients (n = 56) with non-small-cell lung cancer deemed suitable for treatment with nivolumab. MATERIALS & METHODS: This was a multicenter, noninterventional, retrospective data analysis, involving five oncology centers. RESULTS: Patients with prenivolumab NLR values of <5 and ≥5 had respective median overall survival (OS) values of 14.5 and 7.02 months (p = 0.0026). Patients with ≤2 and >2 metastatic sites had median OS values of 11.4 and 6.1 months, respectively (p = 0.0174). A Cox multiple regression model revealed baseline NLR ≥5 as the only variable significantly associated with decreased OS (p < 0.0447). CONCLUSION: Pretreatment elevated NLR values are associated with poor outcomes in patients with recurrent metastatic non-small-cell lung cancer treated with nivolumab.

11.
Cells ; 9(7)2020 07 10.
Article in English | MEDLINE | ID: mdl-32664328

ABSTRACT

High mobility group box 1 (HMGB1) is an extremely versatile protein that is located predominantly in the nucleus of quiescent eukaryotic cells, where it is critically involved in maintaining genomic structure and function. During cellular stress, however, this multifaceted, cytokine-like protein undergoes posttranslational modifications that promote its translocation to the cytosol, from where it is released extracellularly, either actively or passively, according to cell type and stressor. In the extracellular milieu, HMGB1 triggers innate inflammatory responses that may be beneficial or harmful, depending on the magnitude and duration of release of this pro-inflammatory protein at sites of tissue injury. Heightened awareness of the potentially harmful activities of HMGB1, together with a considerable body of innovative, recent research, have revealed that excessive production of HMGB1, resulting from misdirected, chronic inflammatory responses, appears to contribute to all the stages of tumorigenesis. In the setting of established cancers, the production of HMGB1 by tumor cells per se may also exacerbate inflammation-related immunosuppression. These pro-inflammatory mechanisms of HMGB1-orchestrated tumorigenesis, as well as the prognostic potential of detection of elevated expression of this protein in the tumor microenvironment, represent the major thrusts of this review.


Subject(s)
HMGB1 Protein/metabolism , Neoplasms/metabolism , Cytokines/metabolism , Humans , Neoplasms/pathology , Receptor for Advanced Glycation End Products/metabolism , Signal Transduction/physiology , Toll-Like Receptors/metabolism
12.
Molecules ; 25(7)2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32244751

ABSTRACT

Notwithstanding the well-recognized involvement of chronic neutrophilic inflammation in the initiation phase of many types of epithelial cancers, a growing body of evidence has also implicated these cells in the pathogenesis of the later phases of cancer development, specifically progression and spread. In this setting, established tumors have a propensity to induce myelopoiesis and to recruit neutrophils to the tumor microenvironment (TME), where these cells undergo reprogramming and transitioning to myeloid-derived suppressor cells (MDSCs) with a pro-tumorigenic phenotype. In the TME, these MDSCs, via the production of a broad range of mediators, not only attenuate the anti-tumor activity of tumor-infiltrating lymphocytes, but also exclude these cells from the TME. Realization of the pro-tumorigenic activities of MDSCs of neutrophilic origin has resulted in the development of a range of adjunctive strategies targeting the recruitment of these cells and/or the harmful activities of their mediators of immunosuppression. Most of these are in the pre-clinical or very early clinical stages of evaluation. Notable exceptions, however, are several pharmacologic, allosteric inhibitors of neutrophil/MDSC CXCR1/2 receptors. These agents have entered late-stage clinical assessment as adjuncts to either chemotherapy or inhibitory immune checkpoint-targeted therapy in patients with various types of advanced malignancy. The current review updates the origins and identities of MDSCs of neutrophilic origin and their spectrum of immunosuppressive mediators, as well as current and pipeline MDSC-targeted strategies as potential adjuncts to cancer therapies. These sections are preceded by a consideration of the carcinogenic potential of neutrophils.


Subject(s)
Disease Susceptibility , Neoplasms/etiology , Neoplasms/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Animals , Cell Transformation, Neoplastic , Disease Management , Host-Pathogen Interactions , Humans , Immunomodulation , Inflammation/complications , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/drug therapy , Neoplasms/pathology , Neutrophils/drug effects , Oxidation-Reduction , Oxidative Stress , Treatment Outcome
13.
Front Oncol ; 9: 659, 2019.
Article in English | MEDLINE | ID: mdl-31396484

ABSTRACT

Nivolumab (PD-1 inhibitor) and other immune checkpoint inhibitors are used primarily to promote reactivation of anti-tumor immunity. However, due to their generalized immunorestorative properties, these agents may also trigger an unusual spectrum of side-effects termed immune-related adverse events. In the case of the lung, pulmonary infiltrates in patients treated with the anti-PD-1 inhibitors, nivolumab, or pembrolizumab, especially patients with non-small cell lung cancer, can result from immune-related pneumonitis, which, until fairly recently was believed to be of non-infective origin. This, in turn, may result in progression and pseudo-progression of disease. An increasing body of evidence has, however, identified pulmonary tuberculosis as an additional type of anti-PD-1 therapy-associated, immune-related adverse event, seemingly as a consequence of excessive reactivation of immune responsiveness to latent Mycobacterium tuberculosis infection. The current case report describes a 56-year old Caucasian female who presented with microbiologically-confirmed tuberculosis infection while on nivolumab therapy for non-small cell lung cancer. Notably, the patient, seemingly the first described from the African Continent, had not received immunosuppressive therapy prior to the diagnosis of tuberculosis.

14.
BMC Cancer ; 18(1): 917, 2018 Sep 24.
Article in English | MEDLINE | ID: mdl-30249215

ABSTRACT

BACKGROUND: Febrile neutropenia (FN) is a common occurrence during chemotherapy. Granulocyte colony-stimulating factors (G-CSFs) can significantly reduce the risk of FN. International guidelines recommend G-CSF for patients receiving chemotherapy with FN risk of ≥20% or 10% to 20% with defined risk factors. Prophylaxis is not typically recommended for FN risk of < 10%; however, few studies have investigated FN incidence in lower-risk patients in real-world settings and tried to identify higher-risk subgroups. METHODS: This real-world prospective, observational, multinational study aims to estimate the rate of development of FN with a chemotherapy line expected to be associated with a 10% to 20% risk of FN. Eligible patients (> 18 years of age) will have a solid tumour or Hodgkin/non-Hodgkin lymphoma and a planned chemotherapy regimen with expected risk of FN of 10% to 20% (according to published guidelines). Patients will be observed for the duration of the chemotherapy line (first cycle administered without FN prophylaxis). Primary endpoint is incidence of FN after the first chemotherapy cycle. Secondary outcomes include: FN-associated morbidity and mortality; time to first FN occurrence; other FN risk factors and impact of FN on quality of life. A risk model using occurrence of FN as a binary outcome will be developed. Data will be stratified by age, comorbidities and other risk factors. DISCUSSION: This study will provide insight into the real FN risk for common chemotherapy regimens and predictive factors for FN, including patients generally excluded from randomised clinical trials, from which reported FN rates have been variable. This study builds on knowledge of predictive factors from other research and will provide information on patients with 10% to 20% FN risk.


Subject(s)
Antineoplastic Agents/adverse effects , Chemotherapy-Induced Febrile Neutropenia/epidemiology , Clinical Protocols , Neoplasms/complications , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy-Induced Febrile Neutropenia/diagnosis , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Incidence , Neoplasms/drug therapy , Neoplasms/prevention & control , Prognosis , Quality of Life , Research Design , Risk Assessment , Risk Factors
15.
Expert Opin Drug Saf ; 16(6): 697-710, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28460548

ABSTRACT

INTRODUCTION: Five NK-1 RA formulations are commercially available to treat the delayed phase of chemotherapy-induced nausea and vomiting (CINV) occurring between days 2-5 post chemotherapy (aprepitant oral capsule and suspension, fosaprepitant intravenous infusion, netupitant/palonosetron capsules and rolapitant tablet) but no direct comparative studies have been conducted to determine their relative clinical utility. Areas covered: Information on pharmacology and safety of the NK-1 RAs derived from PubMed showed that all bind the NK-1 receptor with high affinity and selectivity. There is substantial variation in the disposition and time course in the body of NK-1 RAs because of the differential effects of hepatic metabolism. Unlike netupitant and rolapitant, aprepitant is metabolized extensively by cytochrome P450 (CYP) 3A4. Aprepitant and netupitant also both inhibit CYP3A4. Consequently, aprepitant not only has a much shorter elimination half-life than netupitant and rolapitant but also a more prolific drug interaction profile. All of the NK-1 RAs are efficacious and safe, and are suitable for use in a range of different patient populations, including those with mild or moderate hepatic or renal impairment. Expert opinion: While discovery of NK-1 RAs represents a major breakthrough in CINV control, further work is needed to improve control of chemotherapy-induced nausea.


Subject(s)
Antiemetics/pharmacology , Nausea/drug therapy , Neurokinin-1 Receptor Antagonists/pharmacology , Vomiting/drug therapy , Animals , Antiemetics/administration & dosage , Antiemetics/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug Interactions , Half-Life , Humans , Nausea/chemically induced , Neurokinin-1 Receptor Antagonists/administration & dosage , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Time Factors , Vomiting/chemically induced
16.
Expert Rev Clin Pharmacol ; 10(1): 17-29, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27894202

ABSTRACT

INTRODUCTION: Chemotherapy-induced nausea and vomiting (CINV) is a debilitating side effect of many cytotoxic chemotherapy regimens. Although sustained antiemetic control across repeated chemotherapy cycles is important for cancer treatment continuation, few studies have investigated the efficacy of antiemetic prophylaxis over multiple chemotherapy cycles. Areas covered: Here we discuss the use of antiemetic hydroxytryptamine type 3 (5-HT3) receptor and neurokinin (NK)-1 receptor antagonists for prevention of CINV, limiting our review to clinical trials in the context of multiple-cycle chemotherapy, with a focus on the NK-1 receptor antagonist rolapitant. 5-HT3 receptor antagonists may be effective in controlling CINV over repeated chemotherapy cycles, but evidence comes primarily from noncomparative studies. NK-1 receptor antagonists provide increased protection against CINV but differences in endpoint selection and methods of analysis preclude meaningful comparisons between agents. Rolapitant shows sustained control of emesis and nausea over multiple cycles of chemotherapy, and compared to other NK-1 receptor antagonists, has a longer half-life and reduced potential for cytochrome P450 3A4-mediated drug-drug interactions. Expert commentary: Trial design should be a key consideration in future studies of CINV therapies, including analytical methods utilized, choice of endpoints, and methods for accounting for nonresponders and patient attrition over multiple cycles of chemotherapy.


Subject(s)
Nausea/prevention & control , Spiro Compounds/therapeutic use , Vomiting/prevention & control , Antiemetics/pharmacology , Antiemetics/therapeutic use , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Drug Interactions , Humans , Nausea/chemically induced , Neoplasms/drug therapy , Neurokinin-1 Receptor Antagonists/therapeutic use , Spiro Compounds/pharmacology , Time Factors , Vomiting/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...