Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
J Health Econ Outcomes Res ; 10(2): 121-131, 2023.
Article in English | MEDLINE | ID: mdl-38093906

ABSTRACT

Background: Long-acting somatostatin analog therapy (LA-SSA) is recommended as first-line therapy for treatment of unresectable or metastatic neuroendocrine tumors (NETs). Understanding treatment sequencing and dosing patterns of LA-SSA is essential for clinical decision-making to provide value-based management of NETs. Objective: To describe treatment patterns of LA-SSA among patients with NETs and subgroups with carcinoid syndrome (CS) in the United States. Methods: This retrospective study utilized claims data from MarketScan® databases to identify patients with NETs and newly treated with LA-SSA between January 1, 2015, and October 31, 2020. Patients were stratified by index LA-SSA (lanreotide and octreotide long-acting release [LAR]). Reported 28-day doses were based on claim fields for days' supply/drug quantity or units of service. Dose escalation was defined as increases in quantity or frequency. Continuous variables, categorical variables, and Kaplan-Meier estimated treatment durations were compared using t-tests, chi-square/Fisher's tests, and log-rank tests, respectively. Results: The study included 241 lanreotide and 521 octreotide LAR patients. Compared with octreotide LAR patients, treatment duration was longer for lanreotide patients (median, 41.3 vs 26.8 months; log-rank p=.004). Fewer lanreotide patients received rescue treatment with short-acting octreotide (7.9% vs 14.4%; p=.011), and a first (6.2% vs 27.3%) and second dose escalation (0.8% vs 5.2%; both p<.05). Among patients with doses reported, fewer lanreotide patients received above-label doses (2.5% [5/202] vs 14.4% [60/416]; p<.001). Among patients who ended treatment during follow-up, fewer lanreotide patients transitioned to another LA-SSA (18.9% [17/90] vs 33.6% [92/274]; p=.008). Similar treatment patterns were observed in CS subgroups. Results for switched treatment patterns were limited due to insufficient sample sizes. Discussion: Real-world treatment patterns of LA-SSA were assessed using more recent administrative claims data. Compared with octreotide LAR patients, lanreotide patients were more likely to remain longer on initial treatment and starting dose without dose escalations and less likely to use rescue treatment and transition to another LA-SSA after discontinuation of the index treatment. Conclusions: Findings from this claims study suggest a potential clinical benefit of lanreotide in NET management.

2.
Curr Oncol Rep ; 24(6): 703-714, 2022 06.
Article in English | MEDLINE | ID: mdl-35254612

ABSTRACT

PURPOSE OF REVIEW: The field of neuroendocrine oncology has changed much since the time of Oberndorfer first described and coined the term carcinoid. The purpose of this review is to summarize recent findings and highlight clinically relevant updates in the management of NENs, particularly those that are practice changing. RECENT FINDINGS: Neuroendocrine tumors (NETs) have replaced carcinoid tumor, for the most part. The classification of neuroendocrine neoplasms (NENs) improved, and the epidemiological understanding of this disease group also expanded with global collaborations and maturation of large tumor registries. Clarity in the utility of some NET biomarkers continues to be evolving. Knowledge of molecular drivers of tumorigenesis increases, and scientific/technological advancements lead the way to multiple drug approvals for the treatment of advanced NETs. The incidence and prevalence of NENs continue to increase, and patients are living longer. Better understanding of molecular drivers and further understanding of the role of immunotherapy in NENs will further elevate the level of care and transform care for all patients with NENs.


Subject(s)
Neuroendocrine Tumors , Humans , Immunotherapy , Incidence , Neuroendocrine Tumors/epidemiology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/therapy , Registries
3.
Endocrine ; 71(2): 502-513, 2021 02.
Article in English | MEDLINE | ID: mdl-33052555

ABSTRACT

PURPOSE: In the phase III CLARINET study (NCT00353496), lanreotide autogel/depot (lanreotide) significantly improved progression-free survival (PFS) vs placebo in patients with non-functioning intestinal or pancreatic neuroendocrine tumours (NETs). The aim of CLARINET open-label extension (OLE) (NCT00842348) was to evaluate long-term safety and efficacy of lanreotide in these patients. METHODS: Patients from the CLARINET study were eligible for the OLE if they had stable disease (irrespective of treatment group) or progressive disease (PD) (placebo-treated patients only). All patients in the OLE received lanreotide 120 mg every 28 days. Computed tomography or magnetic resonance imaging scans were conducted every 6 months and assessed locally for PD (the final scan was also assessed centrally). RESULTS: Overall, 89 patients took part in the OLE (lanreotide, n = 42; placebo, n = 47). Median (range) exposure to lanreotide in patients who received lanreotide in the core study and OLE (LAN-LAN group) was 59.0 (26.0-102.3) months. In this group, the overall incidences of adverse events (AEs) and treatment-related AEs were lower in the OLE than in the core study. Median [95% CI] PFS in the LAN-LAN group was 38.5 [30.9; 59.4] months. In placebo-treated patients with PD at the end of the core study, time to death or subsequent PD during the OLE was 19 [10.1; 26.7] months. CONCLUSIONS: This study provides new evidence on the long-term safety profile and sustained anti-tumour effects of lanreotide autogel/depot in indolent and progressive metastatic intestinal or pancreatic NETs.


Subject(s)
Antineoplastic Agents , Neuroendocrine Tumors , Pancreatic Neoplasms , Antineoplastic Agents/adverse effects , Humans , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Peptides, Cyclic/therapeutic use , Somatostatin/analogs & derivatives
4.
Anticancer Drugs ; 31(3): 216-222, 2020 03.
Article in English | MEDLINE | ID: mdl-31977567

ABSTRACT

Accessible prognostic tools are needed to individualize treatment of neuroendocrine tumors (NETs). Data suggest neutrophil/lymphocyte ratios (NLRs) have prognostic value in some solid tumors, including NETs. In the randomized double-blind CLARINET study (NCT00353496; EudraCT 2005-004904-35), the somatostatin analog lanreotide autogel/depot increased progression-free survival (PFS) compared with placebo in patients with inoperable or metastatic intestinal and pancreatic NETs (grades 1-2, Ki-67 < 10%). The exploratory post-hoc analyses presented here evaluated the prognostic value of NLR in the CLARINET study cohort, in the context of and independently from treatment. Kaplan-Meier PFS plots were generated for patients with available NLR data, in subgroups based on NLR values, and 24-month survival rates were calculated. P values and hazard ratios for prognostic effects were generated using Cox models. 31216222 Baseline characteristics were balanced between lanreotide autogel/depot 120 mg (n = 100) and placebo (n = 101) arms. Irrespective of treatment, raw 24-month PFS rates were comparable across subgroups based on NLR tertiles [37.3% (low), 38.8% (middle), 38.8% (high); n = 67 per group] and NLR cutoff of 4 [38.1% (NLR ≤ 4; n = 176), 40.0% (NLR > 4; n = 25)]. Furthermore, NLRs were not prognostic in Cox models, irrespective of subgroups used. The therapeutic effect of lanreotide autogel/depot 120 mg was independent of NLRs (P > 0.1). These exploratory post-hoc analyses in patients with advanced intestinal and pancreatic NETs contrast with previous data suggesting NLR has prognostic potential in NETs. This may reflect the inclusion of patients with lower-grade tumors or use of higher NLR cutoff values in the current analysis.


Subject(s)
Lymphocytes , Neuroendocrine Tumors/mortality , Neutrophils , Pancreatic Neoplasms/mortality , Aged , Double-Blind Method , Female , Humans , Male , Middle Aged , Neuroendocrine Tumors/blood , Pancreatic Neoplasms/blood , Prognosis , Proportional Hazards Models
5.
Int J Cancer ; 146(7): 1836-1840, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31241762

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare malignancy whose risk factors are unclear. We explored the association of ACC risk with exposure to selected environmental factors, with a focus on cigarette smoking. We conducted a hospital-based case-control study at The University of Texas MD Anderson Cancer Center. Cases (n = 432) patients with ACC treated at MD Anderson, and controls (n = 1,204) were healthy and genetically unrelated spouses of patients at MD Anderson who had cancers not associated with smoking. Information on the subjects' demographic features and selected risk factors was collected using a structured, validated questionnaire and medical records review. Unconditional logistic regression was used to calculate adjusted odds ratios (AORs) via the maximum-likelihood method. Cases had a younger mean (± standard deviation) age than did controls (47.0 ± 0.7 and 60.0 ± 0.3 years, respectively), and the majority of cases were female (60.6%) and non-Hispanic white (82.4%). We found a markedly increased risk of ACC among male cigarette smokers, with an AOR = 1.8 (95% confidence interval [CI] =1.2-2.9), but not among female smokers (AOR = 1.1, 95% CI = 0.7-1.6). Family history of cancer was associated with increased risk of ACC (AOR = 2.8, 95% CI 1.9-4.3) and in both men and women, whereas alcohol consumption was associated with reduced risk in men (AOR = 0.2, 95% CI = 0.1-0.3) but not women (AOR = 0.7, 95% CI = 0.5-1.1). Understanding these risk factors and their underlying mechanisms may help prevent ACC in susceptible individuals and eventually identify new therapeutic options for ACC.


Subject(s)
Adrenocortical Carcinoma/epidemiology , Adrenal Cortex Neoplasms/epidemiology , Adrenal Cortex Neoplasms/etiology , Adrenocortical Carcinoma/etiology , Aged , Case-Control Studies , Female , Hospitalization , Humans , Male , Middle Aged , Odds Ratio , Risk Assessment , Risk Factors
6.
Ann Transl Med ; 7(20): 595, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31807576

ABSTRACT

Immunotherapy is the new frontier in cancer medicine. This manuscript summarizes historical aspect of immunotherapy, particularly its pathway to drug approval as the main therapeutic modality used in clinical medicine. We will discuss the role of immunotherapy in treating various types of cancers and how the treatment landscape once dominated by chemotherapy is rapidly changing.

7.
Clin Adv Hematol Oncol ; 17(11): 630-637, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31851165

ABSTRACT

Cholangiocarcinoma (CCA) encompasses a rare group of malignancies arising from epithelial cells lining the biliary tree that connects the liver and gallbladder to the small intestine. Most patients present with advanced incurable disease that has a poor prognosis, and standard treatment options remain limited. Effective nontoxic treatment options for advanced CCA are needed. Fibroblast growth factors (FGFs) and their fibroblast growth factor receptor (FGFR) pathways are crucial to cellular proliferation, cellular survival, and differentiation of many malignancies, but are especially relevant in CCA. The targeting of FGF/FGFR has become the most promising approach to treating patients with advanced/metastatic CCA. Here we review CCA, and discuss the promise of FGFR-directed therapy in advanced CCA.


Subject(s)
Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Molecular Targeted Therapy , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Clinical Trials as Topic , Humans , Treatment Outcome
8.
Clin Adv Hematol Oncol ; 17(9): 518-523, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31549973

ABSTRACT

Immunotherapy has revolutionized the treatment of various types of cancers in recent years. Since the US Food and Drug Administration approval of the anti-cytotoxic T-lymphocyte-associated antigen 4 agent ipilimumab for late-stage melanoma in 2011, results from multiple clinical trials have proven the benefit of immunotherapy in the treatment of other cancers. However, therapeutic resistance to immunotherapy often develops. This has led investigators to combine immunotherapy with stereotactic body radiation therapy (SBRT) in an attempt to improve outcomes. The benefit of the combination is believed to stem from stimulating and suppressing various immune pathways and is further aided by the abscopal effect, in which tumors respond to radiation therapy even in nonradiated metastatic sites. When combined with immunotherapy, radiation causes the tumor to act much like a vaccine by exposing the tumor antigens to activate the immune response. This article reviews the association between the immune system and cancer, as well as the additional systemic benefit that SBRT can have in patients with advanced-stage malignancies being treated with immunotherapy.


Subject(s)
Immunotherapy , Ipilimumab/therapeutic use , Melanoma/therapy , Radiosurgery , Antigens, Neoplasm/immunology , Humans , Melanoma/immunology , Melanoma/pathology , Neoplasm Staging
9.
Oncologist ; 24(4): 463-474, 2019 04.
Article in English | MEDLINE | ID: mdl-30355775

ABSTRACT

BACKGROUND: Urinary 5-hydroxyindoleacetic acid (5-HIAA) is an established biomarker in neuroendocrine tumors and carcinoid syndrome; however, its role in nonfunctional neuroendocrine tumors is not defined. We present post hoc data on urinary 5-HIAA and plasma chromogranin A (CgA) from the CLARINET study. METHODS: Patients with well- or moderately differentiated, nonfunctioning, locally advanced or metastatic enteropancreatic neuroendocrine tumors were randomized to deep subcutaneous lanreotide depot/autogel 120 mg or placebo once every 28 days for 96 weeks. Tumor response, evaluated centrally (RECIST 1.0), and progression-free survival (PFS) were assessed by treatment and biochemical response, defined as (a) baseline >upper limit of normal (ULN, 41.6 µmol per day 5-HIAA; 98.1 µg/L CgA) and (b) ≥50% decrease from baseline and to ≤ULN value on study. RESULTS: Forty-eight percent (82 of 171; lanreotide, n = 45; placebo, n = 37) and 66% (129 of 195; lanreotide, n = 65; placebo, n = 64) of randomized patients had 5-HIAA and CgA > ULN at baseline. Among patients with >ULN baseline values who did not progress after 96 weeks of treatment, significantly greater reductions in 5-HIAA and CgA were observed in lanreotide-treated versus placebo-treated patients throughout the study (all p < .05). PFS was significantly prolonged among 5-HIAA responders versus nonresponders (median not reached vs. 16.2 months, p < .0001; hazard ratio [HR] = 0.21, 95% confidence interval [CI], 0.09-0.48) and CgA responders versus nonresponders (median not reached vs. 16.2 months, p = .0070; HR = 0.30, 95% CI, 0.12-0.76), regardless of treatment arm. PFS was also significantly prolonged among lanreotide-treated 5-HIAA responders versus nonresponders (p = .0071) but was not significantly different among placebo-treated 5-HIAA responders versus nonresponders. There were no significant differences in PFS between lanreotide-treated CgA responders versus nonresponders or between placebo-treated CgA responders versus nonresponders. CONCLUSIONS: The 5-HIAA findings are noteworthy because they occurred in patients with nonfunctioning enteropancreatic neuroendocrine tumors. Monitoring 5-HIAA and CgA may be useful when treating patients with nonfunctional neuroendocrine tumors. IMPLICATIONS FOR PRACTICE: Current guidelines focus only on the monitoring of 5-hydroxyindoleacetic acid (5-HIAA) in the diagnosis and management of functional neuroendocrine tumors with carcinoid syndrome. The current post hoc analysis of patients with nonfunctional enteropancreatic neuroendocrine tumors in the CLARINET study demonstrated that measuring and following both 5-HIAA and chromogranin A as biomarkers of disease progression may be useful in the management of patients with nonfunctional neuroendocrine tumors.


Subject(s)
Biomarkers, Tumor/analysis , Chromogranin A/blood , Gastrointestinal Neoplasms/secondary , Hydroxyindoleacetic Acid/urine , Neuroendocrine Tumors/secondary , Pancreatic Neoplasms/pathology , Peptides, Cyclic/therapeutic use , Somatostatin/analogs & derivatives , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Female , Follow-Up Studies , Gastrointestinal Neoplasms/blood , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/urine , Humans , International Agencies , Male , Middle Aged , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/urine , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/urine , Prognosis , Retrospective Studies , Somatostatin/therapeutic use , Survival Rate
10.
Endocr Pract ; 2018 Aug 07.
Article in English | MEDLINE | ID: mdl-30084687

ABSTRACT

OBJECTIVE: Neuroendocrine tumors (NETs) are associated with elevated 5-hydroxyindoleacetic acid (5-HIAA) and chromogranin A (CgA) levels. This study aimed to analyze relationships between urinary 5-HIAA and plasma CgA levels and clinical outcomes. METHODS: Centrally assessed biomarker levels and correlations with progression-free survival (PFS) and carcinoid syndrome (CS) symptom control were evaluated in a pooled analysis of CLARINET (96-week randomized, double-blind, placebo-controlled) and ELECT (16-week randomized, double-blind, placebo-controlled, 32-week initial open label and ≥2 year long-term extension open label) studies of adults with NETs, with (ELECT) or without (CLARINET) CS at 97 institutions. Patients were treated with subcutaneous lanreotide depot 120 mg monthly. RESULTS: Of 319 pooled patients, 86% and 95% had baseline 5-HIAA and CgA data, respectively, with 47% and 74% having levels greater than the upper limit of normal (ULN). PFS was longer among patients who experienced a decrease in biomarker levels at week 12, with statistical significance reached in the CgA cohort (not reached vs. 14.4 months; P<.0001). A large proportion (87%) of patients without symptoms of CS in the CLARINET study had detectable levels of 5-HIAA (48% >ULN). In ELECT, patients with CS who received lanreotide and experienced a biochemical response (≥50% decrease from baseline) achieved greater symptom control. CONCLUSION: This pooled analysis of two randomized, placebo-controlled trials demonstrated that 5-HIAA and CgA are secreted as biochemical biomarkers in many patients with NETs, regardless of clinical syndromes. Significant biochemical response was associated with improved clinical outcomes, as measured by improved PFS or improved CS symptom control. ABBREVIATIONS: 5-HIAA = 5-hydroxyindoleacetic acid; CgA = chromogranin A; CI = confidence interval; CLARINET = Controlled Study of Lanreotide Antiproliferative Response in Neuroendocrine Tumors; CS = carcinoid syndrome; ELECT = Evaluation of Lanreotide Depot/Autogel Efficacy and Safety as a Carcinoid Syndrome Treatment; HR = hazard ratio; ITT = intention-to-treat; NET = neuroendocrine tumor; PanNET = pancreatic NET; PFS = progression-free survival; PPI = proton pump inhibitor; SSA = somatostatin analogue; ULN = upper limit of normal.

11.
Oncotarget ; 9(46): 27958-27973, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29963254

ABSTRACT

Chemotherapy-induced diarrhea (CID), with clinical high incidence, adversely affects the efficacy of cancer treatment and patients' quality of life. Our study demonstrates that the citrus flavonoid hesperetin (Hst) has a superior potential as a new agent to prevent and alleviate CID. In the animal model for irinotecan (CPT-11) induced CID, Hst could selectively inhibit intestinal carboxylesterase (CES2) and thus reduce the local conversion of CPT-11 to cytotoxic SN-38 which causes intestinal toxicity. Oral administration of Hst manifested an excellent anti-diarrhea efficacy, prohibiting 80% of severe and 100% of mild diarrhea in the CPT-11 administered tumor-bearing mice. In addition, a significant attenuation of intestinal inflammation contributed to the anti-diarrhea effect of Hst. Moreover, Hst was found to work synergistically with CPT-11 in tumor inhibition by suppressing the tumor's STAT3 activity and recruiting tumoricidal macrophages into the tumor microenvironment. The anti-intestinal inflammation and anti-STAT3 properties of Hst would contribute its broad benefits to the management of diarrhea caused by other chemo or targeted agents, and more importantly, enhance and reinforce the anti-tumor effects of these agents, to improve patient outcomes.

12.
Clinicoecon Outcomes Res ; 9: 495-503, 2017.
Article in English | MEDLINE | ID: mdl-28860831

ABSTRACT

OBJECTIVE: With the introduction of new therapies, hospitals have to plan spending limited resources in a cost-effective manner. To assist in identifying the optimal treatment for patients with locally advanced or metastatic gastroenteropancreatic neuroendocrine tumors, budget impact modeling was used to estimate the financial implications of adoption and diffusion of somatostatin analogs (SSAs). PATIENTS AND METHODS: A hypothetical cohort of 500 gastroenteropancreatic neuroendocrine tumor patients was assessed in an economic model, with the proportion with metastatic disease treated with an SSA estimated using published data. Drug acquisition, preparation, and administration costs were based on national pricing databases and published literature. Octreotide dosing was based on published estimates of real-world data, whereas for lanreotide, real-world dosing was unavailable and we therefore used the highest indicated dosing. Alternative scenarios reflecting the proportion of patients receiving lanreotide or octreotide were considered to estimate the incremental budget impact to the hospital. RESULTS: In the base case, 313 of the initial 500 gastroenteropancreatic neuroendocrine tumor patients were treated with an SSA. The model-predicted per-patient cost was US$83,473 for lanreotide and US$89,673 for octreotide. With a hypothetical increase in lanreotide utilization from 5% to 30% of this population, the annual model-projected hospital costs decreased by US$488,615. When varying the inputs in one-way sensitivity analyses, the results were most sensitive to changes in dosing assumptions. CONCLUSION: Results suggest that factors beyond drug acquisition cost can influence the budget impact to a hospital. When considering preparation and administration time, and real-world dosing, use of lanreotide has the potential to reduce health care expenditures associated with metastatic gastroenteropancreatic neuroendocrine tumor treatments.

13.
Clin Gastroenterol Hepatol ; 15(11): 1791-1799, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28579181

ABSTRACT

BACKGROUND & AIMS: Environmental factors have been identified that affect risk of hepatocellular carcinoma (HCC), but little is known about the effects of sex hormones on liver cancer development or outcome. The authors investigated whether menopause hormone therapy (MHT) affects risk, age at onset, or outcome of HCC. METHODS: We performed a case-control study of 234 female patients treated for HCC at a tertiary medical center and with 282 healthy women (controls) from January 1, 2004 through May 31, 2015. We collected detailed information on environmental exposures, ages of menarche and menopause, hysterectomies, and uses of birth control and MHT. We performed multivariable logistic and Cox regression analyses to determine the independent effects of factors associated with women on risk and clinical outcome in HCC. The primary outcomes were effect of MHT on HCC risk, the relationship between MHT with hepatitis virus infection on HCC development, and effect of MHT on age at HCC onset or survival after diagnosis of HCC. RESULTS: The estimated adjusted odds ratio (AOR) for HCC in women who ever used estrogen was 0.53 (95% confidence interval [CI], 0.32-0.88). This association was supported by the older age of HCC onset among estrogen users (mean, 64.5 ± 0.9 years) vs nonusers (mean 59.2 ± 1.1 years; P = .001) and the reduced risk of HCC among long-term users (more than 5 years) (AOR, 0.36; 95% CI, 0.20-0.63). Users of estrogen also had a reduced risk for hepatitis-associated HCC: AOR for users, 4.37 (95% CI, 1.67-11.44) vs AOR for nonusers, 17.60 (95% CI, 3.88-79.83). Estrogen use reduced risk of death from HCC (hazard ratio, 0.55; 95% CI, 0.40-0.77; P = .01). Median overall survival times were 33.5 months for estrogen users (95% CI, 25.7-41.3 months) and 24.1 months for nonusers (95% CI, 19.02-29.30 months; P = .008). CONCLUSION: In a case-control study of women with HCC vs female control subjects at a single center, we associated use of estrogen MHT with reduced risk of HCC and increased overall survival times of patients with HCC. Further studies are needed to determine the benefits of estrogen therapy for women and patients with HCC, and effects of tumor expression of estrogen receptor.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/epidemiology , Estrogen Replacement Therapy/methods , Liver Neoplasms/drug therapy , Liver Neoplasms/epidemiology , Adult , Aged , Carcinoma, Hepatocellular/mortality , Case-Control Studies , Female , Humans , Incidence , Liver Neoplasms/mortality , Middle Aged , Risk Assessment , Survival Analysis , Treatment Outcome
14.
Clin Adv Hematol Oncol ; 15 Suppl 4(4): 1-24, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28654022

ABSTRACT

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are rare malignancies that originate in the gastrointestinal system. GEP-NETs are typically indolent, but tumors known as "functional" secrete hormones that can lead to a complex of symptoms, including flushing, diarrhea, bronchospasm, and valvular heart disease. Management of patients with GEP-NETs requires a multidisciplinary approach, as treatment modalities include surgery, radiology, and pharmacotherapy. The available pharmacologic agents have increased in recently, and now include cytotoxic chemotherapies, somatostatin analogues, multitargeted tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors, and radioisotopic radiotherapies. The optimal sequencing of treatments is unknown. Advances in the management of GEP-NETs have been based on the results of recently completed clinical trials that have shown improvement in disease outcome and symptom management. The amount of positive data that has emerged from these studies is unprecedented in the GEP-NETs field. At the 2017 American Society of Clinical Oncology Gastrointestinal Cancers Symposium, several abstracts provided subanalyses of previous trials and new data for emerging treatments. Management will likely evolve as these therapies are incorporated into clinical care.


Subject(s)
Antineoplastic Agents/therapeutic use , Intestinal Neoplasms/drug therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Clinical Trials as Topic , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/pathology , Humans , Intestinal Neoplasms/pathology , Neuroendocrine Tumors/pathology , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/pathology , Peptides, Cyclic/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Stomach Neoplasms/pathology
17.
Leuk Lymphoma ; 58(9): 1-11, 2017 09.
Article in English | MEDLINE | ID: mdl-28140714

ABSTRACT

Hyperleukocytosis in patients with acute myeloid leukemia (AML) can lead to leukostasis, which if left untreated, has a high mortality. While prompt cytoreductive chemotherapy is essential, treatment with leukapheresis is controversial. This study investigated the outcomes of patients with hyperleukocytosis who received leukapheresis. From 5596 encounters of patients with leukemia seen at Houston Methodist Hospital, we identified 26 patients who had newly diagnosed AML, WBC >50,000/µL, and received leukapheresis. We matched 26 patients who had similar baseline characteristics but did not receive leukapheresis. The primary endpoint was to compare the 28-day mortality rates between the treatment and the control groups. Secondary endpoints were 6-month, 1-year, and 2-year mortality rates. Using multivariate logistic regression analysis, leukapheresis was associated with significantly lower 28-day mortality rate (30.8% vs. 57.7%, p = .022). There was, however, no difference in long-term mortality rate. Our study demonstrates the short-term mortality benefit of using leukapheresis in AML patients presenting with hyperleukocytosis.


Subject(s)
Leukapheresis , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Case-Control Studies , Female , Humans , Leukapheresis/methods , Leukemia, Myeloid, Acute/pathology , Leukocyte Count , Male , Middle Aged , Registries , Retrospective Studies
18.
Clin J Oncol Nurs ; 20(6): E139-E146, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27857269

ABSTRACT

BACKGROUND: Somatostatin analogs (SSAs) are a mainstay therapy for the treatment of carcinoid syndrome associated with neuroendocrine tumors (NETs). They are effective for a range of gastroenteropancreatic NETs (GEP-NETs). Lanreotide depot (Somatuline®) is an SSA that is approved for the treatment of GEP-NETs to improve progression-free survival (PFS). OBJECTIVES: The article reviews the efficacy, safety, and administration of lanreotide depot and relates those attributes to considerations and preferences of oncology nurses and their patients. METHODS: A review of the literature on the use of lanreotide for the treatment of NETs and carcinoid syndrome was conducted. In addition, the literature on drug delivery and routes of administration was surveyed to provide context for comparative studies related to clinical and patient preferences. FINDINGS: Lanreotide depot prolongs PFS and is well tolerated by patients who expressed satisfaction in the ability to control symptoms related to carcinoid syndrome. Nurses cited several benefits to using lanreotide depot in the clinical setting, including more time saved to address other patient care issues. Attributes of lanreotide depot-including its efficacy, safety and tolerability, dosing and administration, and cost-may contribute to healthcare decisions regarding the treatment and management of NETs.


Subject(s)
Intestinal Neoplasms/drug therapy , Intestinal Neoplasms/mortality , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/mortality , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Peptides, Cyclic/therapeutic use , Somatostatin/analogs & derivatives , Stomach Neoplasms/drug therapy , Stomach Neoplasms/mortality , Antineoplastic Agents/administration & dosage , Clinical Trials, Phase II as Topic , Clinical Trials, Phase III as Topic , Delayed-Action Preparations , Disease-Free Survival , Female , Humans , Injections, Intramuscular , Intestinal Neoplasms/diagnosis , Intestinal Neoplasms/nursing , Male , Neuroendocrine Tumors/diagnosis , Neuroendocrine Tumors/nursing , Neuroendocrine Tumors/pathology , Oncology Nursing/methods , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/nursing , Patient Preference , Prognosis , Somatostatin/therapeutic use , Stomach Neoplasms/diagnosis , Stomach Neoplasms/nursing , Time Factors , Treatment Outcome
19.
J Gastrointest Cancer ; 47(4): 366-374, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27619395

ABSTRACT

PURPOSE: Peptide drugs for antineoplastic therapies usually have low oral bioavailability and short in vivo half-lives, requiring less preferred delivery methods. Lanreotide depot is a sustained-release somatostatin analog (SSA) formulation produced via an innovative peptide self-assembly method. Lanreotide is approved in the USA and Europe to improve progression-free survival (PFS) in patients with unresectable gastroenteropancreatic neuroendocrine tumors (GEP-NETs) and also approved in Europe for symptom control in carcinoid syndrome associated with GEP-NETs. This review discusses how the distinct molecule and formulation of lanreotide depot provide advantages to patients and health care providers, as well as the most recent clinical evidence demonstrating the safety and efficacy of lanreotide depot in inhibiting tumor growth and controlling hormonal symptoms in GEP-NETs. METHODOLOGY AND RESULTS: The lanreotide depot formulation confers a remarkable pharmacokinetic profile with no excipients, comprised only of lanreotide acetate and water. Of note, lanreotide depot constitutes an example for peptide self-assembly based formulations, providing insights that could help future development of sustained-release formulations of other antineoplastic peptides. Most patients with GEP-NETs will present with inoperable or incurable disease; thus, medical management for symptoms and tumor control plays a crucial role. Recent long-term clinical studies have demonstrated that lanreotide depot is well tolerated, prolongs PFS in GEP-NET patients, and significantly reduces symptoms related to carcinoid syndrome. CONCLUSIONS: The unique depot formulation and delivery method of lanreotide confer advantages in the treatment of metastatic GEP-NETs, contributing to improvements in NET-related symptoms and PFS without reducing quality of life in this patient population.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoid Tumor/drug therapy , Carcinoma, Neuroendocrine/drug therapy , Peptides, Cyclic/therapeutic use , Somatostatin/analogs & derivatives , Antineoplastic Agents/administration & dosage , Carcinoid Tumor/pathology , Carcinoma, Neuroendocrine/pathology , Female , Humans , Male , Peptides, Cyclic/administration & dosage , Somatostatin/administration & dosage , Somatostatin/therapeutic use
20.
Cancer Med ; 5(10): 2953-2964, 2016 10.
Article in English | MEDLINE | ID: mdl-27539383

ABSTRACT

Significant advances have been made toward understanding the biology of neuroendocrine tumors (NET) in terms of defining prognosis and improving clinical management; however, many unmet needs remain. The treatment landscape for NET has evolved, with the approval of the targeted agents everolimus and sunitinib for the treatment of advanced pancreatic NET in 2011 followed by the approval of everolimus for the treatment of advanced nonfunctional gastrointestinal and lung NET in 2016. Mammalian target of rapamycin (mTOR) and components of the mTOR pathway play pivotal roles in NET pathogenesis. Effects of the mTOR inhibitor everolimus have been well documented in preclinical and clinical studies, both as monotherapy and combination therapy. mTOR inhibition as backbone therapy within the NET treatment landscape is a focus of continuing research, which includes evaluation of the growing armamentarium of approved and investigational agents as potential combination partners. Data evaluating the clinical benefits of agents targeting mTOR and related pathways (alone and in combination) in the treatment of patients with NET continue to increase. Many of the findings to date are encouraging.


Subject(s)
Antineoplastic Agents/therapeutic use , Neuroendocrine Tumors/drug therapy , Protein Kinase Inhibitors/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Drug Screening Assays, Antitumor , Drug Therapy, Combination , Everolimus/pharmacology , Everolimus/therapeutic use , Humans , Indoles/pharmacology , Indoles/therapeutic use , Neuroendocrine Tumors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Pyrroles/therapeutic use , Signal Transduction/drug effects , Sunitinib , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...