Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pediatr Blood Cancer ; 71(8): e31117, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38804882

ABSTRACT

BACKGROUND: Aromatase inhibitors (AI) may improve height in short stature conditions; however, the effect in childhood cancer survivors (CCS) is unknown. We assessed final adult height (FAH) in CCS treated with AI and GH compared with those treated with GH alone. METHODS: Retrospective cohort study of GH-deficient male CCS treated between 2007 and 2023. FAH was noted as the height at the fusion of growth plates or 18 years of age. Multivariable linear regression was used to examine treatment association with FAH, adjusting for other risk factors. RESULTS: Ninety-two patients were included; 70 were treated with GH and 22 with combination AI/GH. The mean age at GH initiation did not differ between groups. The mean age at AI initiation was 13.7 ± 1.9 years. A greater proportion of patients in the AI/GH group were treated with stem cell transplantation, abdominal radiation, total body irradiation, and cis-retinoic acid (p < .01). Multivariable linear regression demonstrated no significant treatment association with FAH Z-score (ß = 0.04, 95% CI: -0.9 to 0.9). History of spinal radiation (ß = -0.93, 95% CI: -1.7 to -0.2), lower starting height Z-score (ß = -0.8, 95% CI: -1.2 to -0.4), and greater difference between bone age and chronological age (ß = -0.3, 95% CI: -0.5 to -0.07) were associated with lower FAH Z-score. CONCLUSIONS: Adjuvant AI was not associated with increased FAH in male CCS compared with GH monotherapy. Future work is needed to determine the optimal adjunctive treatment to maximize FAH for this population.


Subject(s)
Aromatase Inhibitors , Body Height , Cancer Survivors , Human Growth Hormone , Neoplasms , Humans , Male , Aromatase Inhibitors/therapeutic use , Retrospective Studies , Body Height/drug effects , Adolescent , Human Growth Hormone/deficiency , Child , Neoplasms/drug therapy , Follow-Up Studies , Growth Disorders/drug therapy , Growth Disorders/etiology , Growth Disorders/pathology , Adult , Prognosis , Chemotherapy, Adjuvant
3.
Pediatr Blood Cancer ; 69(7): e29505, 2022 07.
Article in English | MEDLINE | ID: mdl-34931744

ABSTRACT

BACKGROUND: Chemotherapy regimens containing glucocorticoids and pegaspargase are associated with hyperglycemia; however, the pattern and underlying risk factors are not well characterized. We determined the pattern of hyperglycemia and associated factors in children with acute lymphoblastic leukemia (ALL) receiving glucocorticoids and pegaspargase during induction. METHODS: Retrospective analysis of patients treated between 2010 and 2020 at a single institution. Pretreatment data, glucose values, and insulin regimens were abstracted from the record. Hyperglycemia was defined as two or more random glucose measurements ≥200 mg/dl. Analyses of demographic and clinical factors were conducted with logistic regression. RESULTS: Two hundred thirteen patients, median age 6 years (range 1.0-18.9 years), 47% female, were included. The prevalence of hyperglycemia was 23% (n = 48). Mean glucose levels peaked 3 days following administration of pegaspargase. In multivariable analysis, age ≥10 years (odds ratio [OR] 6.2, 95% confidence interval [CI]: 2.9-13.4), female sex (OR 2.7, 95% CI: 1.2-6.2), and family history of diabetes (OR 3.2, 95% CI: 1.4-7.3) were predictive of hyperglycemia. Age ≥10 years (OR 19.4, 95% CI: 5.5-68.4), family history of diabetes (OR 8.2, 95% CI: 2.7-25.3), and higher body mass index (BMI) (OR 1.8, 95% CI: 1.1-2.9) were associated with insulin treatment. CONCLUSIONS: Onset of hyperglycemia in children receiving induction chemotherapy for ALL is temporally linked to administration of pegaspargase. Older age, female sex, and family history of diabetes are predictive of hyperglycemia during induction; older age, family history of diabetes, and higher BMI are associated with insulin treatment. Frequent glucose monitoring is indicated during induction therapy for ALL.


Subject(s)
Diabetes Mellitus , Hyperglycemia , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adolescent , Asparaginase/adverse effects , Blood Glucose , Blood Glucose Self-Monitoring , Child , Child, Preschool , Diabetes Mellitus/epidemiology , Female , Glucocorticoids/therapeutic use , Humans , Hyperglycemia/chemically induced , Hyperglycemia/complications , Hyperglycemia/epidemiology , Induction Chemotherapy , Infant , Insulin , Male , Polyethylene Glycols/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Retrospective Studies
4.
Front Endocrinol (Lausanne) ; 12: 745932, 2021.
Article in English | MEDLINE | ID: mdl-34745010

ABSTRACT

Growth hormone (GH) deficiency is a common pituitary hormone deficiency in childhood cancer survivors (CCS). The identification, diagnosis, and treatment of those individuals at risk are important in order to minimize associated morbidities that can be ameliorated by treatment with recombinant human GH therapy. However, GH and insulin-like growth factor-I have been implicated in tumorigenesis, so there has been concern over the use of GH therapy in patients with a history of malignancy. Reassuringly, GH therapy has not been shown to increase risk of tumor recurrence. These patients have an increased risk for development of meningiomas, but this may be related to their history of cranial irradiation rather than to GH therapy. In this review, we detail the CCS who are at risk for GHD and the existing evidence on the safety profile of GH therapy in this patient population.


Subject(s)
Brain Neoplasms , Cancer Survivors , Human Growth Hormone/deficiency , Adult , Brain Neoplasms/epidemiology , Brain Neoplasms/therapy , Cancer Survivors/statistics & numerical data , Child , Cranial Irradiation/adverse effects , Growth Disorders/drug therapy , Growth Disorders/epidemiology , Growth Disorders/etiology , Growth Disorders/metabolism , Hormone Replacement Therapy , Human Growth Hormone/therapeutic use , Humans , Hypopituitarism/drug therapy , Hypopituitarism/epidemiology , Hypopituitarism/etiology , Hypopituitarism/metabolism , Insulin-Like Growth Factor I/metabolism , Risk Factors
5.
J Chem Biol ; 10(3): 129-141, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28684999

ABSTRACT

Studies indicate that elevated interleukin-6 (IL-6) levels engage IL6Rα-gp130 receptor complexes to activate signal transducer and activator of transcription 3 (STAT3) that is hyperactivated in many cancers including head and neck squamous cell carcinoma (HNSCC). Our previous HCS campaign identified several hits that selectively blocked IL-6-induced STAT3 activation. This study describes our investigation of the mechanism(s) of action of three of the four chemical series that progressed to lead activities: a triazolothiadiazine (864669), amino alcohol (856350), and an oxazole-piperazine (4248543). We demonstrated that all three blocked IL-6-induced upregulation of the cyclin D1 and Bcl-XL STAT3 target genes. None of the compounds exhibited direct binding interactions with STAT3 in surface plasmon resonance (SPR) binding assays; neither did they inhibit the recruitment and binding of a phospho-tyrosine-gp130 peptide to STAT3 in a fluorescence polarization assay. Furthermore, they exhibited little or no inhibition in a panel of 83 cancer-associated in vitro kinase profiling assays, including lack of inhibition of IL-6-induced Janus kinase (JAK 1, 2, and 3) activation. Further, 864669 and 4248543 selectively inhibited IL-6-induced STAT3 activation but not that induced by oncostatin M (OSM). The compounds 864669 and 4248543 abrogated IL-6-induced phosphorylation of the gp130 signaling subunit (phospho-gp130Y905) of the IL-6-receptor complex in HNSCC cell lines which generate docking sites for the SH2 domains of STAT3. Our data indicate that 864669 and 4248543 block IL-6-induced STAT activation by interfering with the recruitment, assembly, or activation of the hexamer-activated IL-6/IL-6Rα/gp130 signaling complex that occurs after IL-6 binding to IL-6Rα subunits.

6.
Pain Rep ; 2(3): e596, 2017 May.
Article in English | MEDLINE | ID: mdl-29392212

ABSTRACT

INTRODUCTION: Chronic pain is a common and debilitating complication following breast surgery. One of the most challenging for treatment is the neuropathic pain condition, postmastectomy pain syndrome (PMPS). Gabapentin is a pharmacotherapy for neuropathic pain disorders; however, its once-daily, gastroretentive formulation, Gralise, has not been evaluated in PMPS. OBJECTIVE: To evaluate the safety and effectiveness of Gralise in patients with moderate-to-severe PMPS. METHODS: The primary effectiveness endpoint was a change in the worst pain intensity score from baseline to completion of 8 weeks of Gralise therapy. The secondary endpoints included the change in mood, coping behavior, sleep, and function. Sensitivity to experimental stimuli was tested before and after treatment via quantitative sensory testing. The incidence and type of adverse event were used to evaluate the safety and tolerability of Gralise. RESULTS: Twenty-one patients with confirmed moderate-to-severe PMPS were enrolled. Nineteen of 21 (90.5%) patients completed the 8-week treatment with Gralise. A significant positive change was found in pain intensity, pain impact, and sleep. There was no change in sensory testing scores. Of total, 63.16% of patients reported reduction in present pain, 78.95% in average pain, 89.47% in worst pain, and 84.21% in overall pain severity at posttreatment visit. No significant adverse effects were noted in the study. LIMITATIONS: Variation in type of breast surgery, small sample size, lack of placebo control. CONCLUSION: There was a significant improvement in pain and sleep, and Gralise was well tolerated in patients with PMPS. Further investigation is warranted.

7.
Bioorg Med Chem Lett ; 26(15): 3581-5, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27381083

ABSTRACT

Structure-activity relationship studies of a 1,2,4-triazolo-[3,4-b]thiadiazine scaffold, identified in an HTS campaign for selective STAT3 pathway inhibitors, determined that a pyrazole group and specific aryl substitution on the thiadiazine were necessary for activity. Improvements in potency and metabolic stability were accomplished by the introduction of an α-methyl group on the thiadiazine. Optimized compounds exhibited anti-proliferative activity, reduction of phosphorylated STAT3 levels and effects on STAT3 target genes. These compounds represent a starting point for further drug discovery efforts targeting the STAT3 pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Pyrazoles/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Thiadiazines/pharmacology , Triazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Pyrazoles/chemistry , STAT3 Transcription Factor/metabolism , Structure-Activity Relationship , Thiadiazines/chemical synthesis , Thiadiazines/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry
8.
Clin Cancer Res ; 21(20): 4597-606, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26138066

ABSTRACT

PURPOSE: In other cancer types, HPV infection has been reported to coincide with overexpression of HER2 (ERBB2) and HER3 (ERBB3); however, the association between HER2 or HER3 expression and dimer formation in HNSCC has not been reported. Overexpression of HER2 and HER3 may contribute to resistance to EGFR inhibitors, including cetuximab, although the contribution of HPV in modulating cetuximab response remains unknown. Determination of heterodimerization of HER receptors is challenging and has not been reported in HNSCC. The present study aimed to determine the expression of HER proteins in HPV(+) versus HPV(-) HNSCC tumors using a proximity-based protein expression assay (VeraTag), and to determine the efficacy of HER-targeting agents in HPV(+) and HPV(-) HNSCC cell lines. EXPERIMENTAL DESIGN: Expression of total HER1, HER2, and HER3, p95HER2, p-HER3, HER1:HER1 homodimers, HER2:HER3 heterodimers, and the HER3-PI3K complex in 88 HNSCC was determined using VeraTag, including 33 baseline tumors from individuals treated in a trial including cetuximab. Inhibition of cell growth and protein activation with cetuximab and afatinib was compared in HPV(+) and HPV(-) cetuximab-resistant cell lines. RESULTS: Expression of total HER2, total HER3, HER2:HER3 heterodimers, and the HER3:PI3K complex were significantly elevated in HPV(+) HNSCC. Total EGFR was significantly increased in HPV(-) HNSCC where VeraTag assay results correlated with IHC. Afatinib significantly inhibited cell growth when compared with cetuximab in the HPV(+) and HPV(-) cetuximab-resistant HNSCC cell lines. CONCLUSIONS: These findings suggest that agents targeting multiple HER proteins may be effective in the setting of HPV(+) HNSCC and/or cetuximab resistance.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Papillomaviridae/pathogenicity , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Cetuximab/therapeutic use , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/metabolism , Female , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/virology , Humans , Male , Middle Aged , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Phosphatidylinositol 3-Kinases/metabolism , Protein Multimerization/drug effects , Squamous Cell Carcinoma of Head and Neck , Young Adult
9.
Neoplasia ; 17(3): 256-64, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25810010

ABSTRACT

Aberrant activation of the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) 3 has been implicated in cell proliferation and survival of many cancers including head and neck squamous cell carcinoma (HNSCC). AZD1480, an orally active pharmacologic inhibitor of JAK1/JAK2, has been tested in several cancer models. In the present study, the in vitro and in vivo effects of AZD1480 were evaluated in HNSCC preclinical models to test the potential use of JAK kinase inhibition for HNSCC therapy. AZD1480 treatment decreased HNSCC proliferation in HNSCC cell lines with half maximal effective concentration (EC50) values ranging from 0.9 to 4 µM in conjunction with reduction of pSTAT3(Tyr705) expression. In vivo antitumor efficacy of AZD1480 was demonstrated in patient-derived xenograft (PDX) models derived from two independent HNSCC tumors. Oral administration of AZD1480 reduced tumor growth in conjunction with decreased pSTAT3(Tyr705) expression that was observed in both PDX models. These findings suggest that the JAK1/2 inhibitors abrogate STAT3 signaling and may be effective in HNSCC treatment approaches.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Janus Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Dosage , Gene Expression , Head and Neck Neoplasms/genetics , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Mice , Phosphorylation , Pyrazoles/pharmacology , Pyrimidines/pharmacology , RNA, Messenger/genetics , Signal Transduction/drug effects , Squamous Cell Carcinoma of Head and Neck , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 21(3): 526-33, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25424855

ABSTRACT

The majority of patients with head and neck squamous cell carcinoma (HNSCC) present with advanced-stage disease. The current standard of care is surgery followed by adjuvant radiotherapy with or without chemotherapy or chemoradiation alone. The addition of cetuximab for the treatment of patients with locally advanced or recurrent/metastatic HNSCC has improved overall survival and locoregional control; however, responses are often modest, and treatment resistance is common. A variety of therapeutic strategies are being explored to overcome cetuximab resistance by blocking candidate proteins implicated in resistance mechanisms such as HER2. Several HER2 inhibitors are in clinical development for HNSCC, and HER2-targeted therapy has been approved for several cancers. This review focuses on the biology of HER2, its role in cancer development, and the rationale for clinical investigation of HER2 targeting in HNSCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Molecular Targeted Therapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/pathology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Head and Neck Neoplasms/pathology , Humans , Signal Transduction , Squamous Cell Carcinoma of Head and Neck
SELECTION OF CITATIONS
SEARCH DETAIL
...