Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Pediatr Blood Cancer ; 70(5): e30217, 2023 05.
Article in English | MEDLINE | ID: mdl-36772891

ABSTRACT

Monoclonal antibodies (mAbs) targeting disialoganglioside 2 (GD2) are an important treatment advance for high-risk neuroblastoma, including in patients with refractory or relapsed disease. Dinutuximab and dinutuximab beta are administered for ≥8 hours (and up to 10 days for dinutuximab beta), whereas naxitamab is administered over 0.5 to 2 hours as tolerated. As acute pain is a class effect of anti-GD2 mAbs, effective pain management is crucial to successful treatment. Here, we provide an overview of current pain-management strategies for anti-GD2 mAb infusions, with a focus on strategies suitable for naxitamab infusions, which cause a more rapid onset of often severe pain. We discuss opioid analgesics, ketamine, gabapentin, and other similar agents and nonpharmacologic approaches. Potential future pain-management options are also discussed, in addition to the use of sedatives to reduce the anxiety that may be associated with infusion-related pain. In this expert consensus paper, specific guidance for pain management during naxitamab infusions is provided, as these infusions are administered over 0.5 to 2 hours and may not need overnight hospitalization based on the physician's assessment, and require rapid-onset analgesia options suitable for potential outpatient administration.


Subject(s)
Antineoplastic Agents , Neuroblastoma , Humans , Antineoplastic Agents/therapeutic use , Consensus , Gangliosides , Immunotherapy , Neuroblastoma/drug therapy , Pain/drug therapy , Pain/etiology , Pain/prevention & control , Pain Management
2.
J Pediatr Hematol Oncol ; 45(1): e124-e125, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36598966

ABSTRACT

This case describes an unusual presentation of a young adult with a very late recurrence of stage MS neuroblastoma over 20 years after initial diagnosis. Tumor histology at relapse demonstrated ganglioneuromatous foci within her undifferentiated tumor. In combination with evidence of altered catecholamine metabolism, it proposes a case for dedifferentiation of unresected ganglioneuromatous lesions as the etiology of her recurrence of the disease. An additional, compelling component of the case is the overall positive treatment response of the patient with relapsed neuroblastoma despite the poor prognostic factors of late relapse and adult age.


Subject(s)
Ganglioneuroma , Neuroblastoma , Female , Humans , Young Adult , Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Chronic Disease , Recurrence
3.
Int J Surg Pathol ; 30(6): 706-715, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35164578

ABSTRACT

We describe two poorly differentiated, non-myofibroblastic (SMA-, S100+, CD34±), spindle cell neoplasms with immunohistochemical positivity for ALK and with ALK gene rearrangements leading to PLEKHH2::ALK and CLTC::ALK fusions, respectively. ALK protein overexpression and/or gene fusions should be evaluated in poorly differentiated spindle cell neoplasms, even when there is an absence of a myofibroblastic phenotype. A positive ALK evaluation has therapeutic implications as both tumors responded to single-agent treatment with the tyrosine kinase inhibitor crizotinib.


Subject(s)
Myofibroblasts , Protein Kinase Inhibitors , Anaplastic Lymphoma Kinase/genetics , Crizotinib/therapeutic use , Gene Rearrangement , Humans , Myofibroblasts/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
4.
Med ; 2(4): 395-410, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33860280

ABSTRACT

Background: Survival among children with high-risk solid tumors remains poor. Reprogrammed metabolism promotes tumor growth and may contain therapeutic liabilities. Tumor metabolism has been assessed in adults using intra-operative 13C-glucose infusions. Pediatric tumors differ from adult cancers in their low mutational burden and derivation from embryonic tissues. Here we used 13C infusions to examine tumor metabolism in children, comparing phenotypes among tumor types and between childhood and adult cancers. Methods: Patients recruited to study NCT03686566 received an intra-operative infusion of [U-13C]glucose during tumor resection to evaluate central carbon pathways in the tumor, with concurrent metabolomics to provide a broad overview of metabolism. Differential characteristics were determined using multiple comparison tests and mixed effect analyses. Findings: We studied 23 tumors from 22 patients. All tumors analyzed by [U-13C]glucose contained labeling in glycolytic and tricarboxylic acid (TCA) cycle intermediates. Labeling in the TCA cycle indicated activity of pyruvate dehydrogenase (PDH) and pyruvate carboxylase (PC), with PDH predominating. Neuroblastomas had high lactate labeling relative to other childhood cancers and lung cancer, and were distinguished by abundant tyrosine catabolites consistent with catecholamine synthesis. Conclusions: Intra-operative [U13C]glucose infusions are safe and informative in pediatric cancer. Tumors of various histologies use glycolysis and oxidative metabolism, with subtype-selective differences evident from this small cohort. Expanding this cohort may uncover predictive biomarkers and therapeutic targets from tumor metabolism. Funding: N.C.I grants to P.L. (R21CA220090-01A1) and R.J.D. (R35CA22044901); H.H.M.I. funding to R.J.D.; Children's Clinical Research Advisory Committee funding to K.J.


Subject(s)
Glycolysis , Neoplasms , Child , Glucose/metabolism , Humans , Isotopes , Oxidative Stress
5.
CA Cancer J Clin ; 71(4): 315-332, 2021 07.
Article in English | MEDLINE | ID: mdl-33793968

ABSTRACT

Although significant improvements have been made in the outcomes of children with cancer, the pace of improvement has slowed in recent years as the limits of therapy intensification may have been reached for many pediatric cancers. Furthermore, with increasing numbers of pediatric cancer survivors, the long-term side effects of treatment have become increasingly apparent. Therefore, attention has shifted to the use of molecularly targeted agents and immunotherapies to improve the outcomes of children who are not cured by traditional cytotoxic chemotherapies and to decrease exposure to cytotoxic chemotherapy and reduce late effects. This review describes the recent progress in the treatment of children with cancer, focusing in particular on diseases in which targeted and immunotherapeutic agents have made an impact.


Subject(s)
Medical Oncology/trends , Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Child , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics
6.
Am Soc Clin Oncol Educ Book ; 40: 1-10, 2020 May.
Article in English | MEDLINE | ID: mdl-32412804

ABSTRACT

Patients and their families, physicians, drug companies, and regulatory agencies have common goals: to find effective therapies for life-threatening conditions. In oncology, the lines between clinical research and treatment are often blurred; parents and physicians of patients who have exhausted standard-of-care treatments and cannot participate in a clinical trial are likely to consider seeking compassionate use access to investigational drugs; however, knowledge and perspectives about compassionate use may differ among these groups. There are unique considerations associated with providing compassionate use to children diagnosed with cancer, including evaluation for potential developmental toxicities, the need for pediatric-specific dosing and formulations, informed consent, and, when appropriate, patient assent. Positive impacts of providing access to investigational therapies to children include potential treatment benefits to patients who obtain investigational agents as well as benefits to future patients if data from expanded access support drug development for childhood cancer. Challenges for physicians seeking compassionate use access to investigational drugs for their patients include obtaining the drug sponsor's agreement to provide the investigational drug as well as lack of knowledge about the process and regulatory requirements. Clinical trials in oncology provide the possibility of therapeutic benefit for pediatric patients; when feasible and warranted, these benefits should also be available to patients on a compassionate use basis outside of trials.


Subject(s)
Compassionate Use Trials , Drugs, Investigational/therapeutic use , Neoplasms/drug therapy , Child , Humans
8.
Int J Radiat Oncol Biol Phys ; 94(4): 800-7, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26972653

ABSTRACT

OBJECTIVES: The majority of childhood cancer patients now achieve long-term survival, but the treatments that cured their malignancy often put them at risk of adverse health outcomes years later. New cancers are among the most serious of these late effects. The aims of this review are to compare and contrast radiation dose-response relationships for new solid cancers in a large cohort of childhood cancer survivors and to discuss interactions among treatment and host factors. METHODS: This review is based on previously published site-specific analyses for subsequent primary cancers of the brain, breast, thyroid gland, bone and soft tissue, salivary glands, and skin among 12,268 5-year childhood cancer survivors in the Childhood Cancer Survivor Study. Analyses included tumor site-specific, individual radiation dose reconstruction based on radiation therapy records. Radiation-related second cancer risks were estimated using conditional logistic or Poisson regression models for excess relative risk (ERR). RESULTS: Linear dose-response relationships over a wide range of radiation dose (0-50 Gy) were seen for all cancer sites except the thyroid gland. The steepest slopes occurred for sarcoma, meningioma, and nonmelanoma skin cancer (ERR/Gy > 1.00), with glioma and cancers of the breast and salivary glands forming a second group (ERR/Gy = 0.27-0.36). The relative risk for thyroid cancer increased up to 15-20 Gy and then decreased with increasing dose. The risk of thyroid cancer also was positively associated with chemotherapy, but the chemotherapy effect was not seen among those who also received very high doses of radiation to the thyroid. The excess risk of radiation-related breast cancer was sharply reduced among women who received 5 Gy or more to the ovaries. CONCLUSIONS: The results suggest that the effect of high-dose irradiation is consistent with a linear dose-response for most organs, but they also reveal important organ-specific and host-specific differences in susceptibility and interactions between different aspects of treatment.


Subject(s)
Dose-Response Relationship, Radiation , Neoplasms, Radiation-Induced , Neoplasms, Second Primary/etiology , Adolescent , Age Factors , Antineoplastic Agents/adverse effects , Child , Child, Preschool , Female , Humans , Infant , Male , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/radiotherapy , Radiotherapy Dosage , Sex Factors , Survivors , Young Adult
9.
Gerontol Geriatr Med ; 2: 2333721416665523, 2016.
Article in English | MEDLINE | ID: mdl-28680937

ABSTRACT

Objective: Where swallowing difficulties are chronic or progressive, or a patient is palliative, tube feeding is often not deemed appropriate. Instead, patients continue to eat and drink despite the risks of pneumonia and death. There is currently little evidence to guide clinical practice in this field often termed "risk feeding." This qualitative study investigated staff, patient, and family member perceptions of risk feeding practices in one New Zealand hospital. Method: Twenty-nine staff members and six patients and/or their family were interviewed. Results: Thematic analysis revealed four global themes: supporting practice, communication, complexity of feeding decisions, and patient and family-centered care. Staff described limited education and organizational policy around risk feeding decisions. Communication was considered a major factor in the success. Conclusion: Feeding decisions are complex in the hospital environment. The themes identified in this study provide a foundation for hospital guideline development and implementation.

10.
Pediatr Blood Cancer ; 62(6): 1048-54, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25662999

ABSTRACT

BACKGROUND: FLT3/ITD is associated with poor outcomes in adult and pediatric acute myeloid leukemia (AML). Allogeneic hematopoietic stem cell transplantation (HSCT) can improve cure rates, however relapse is still common. Recent studies demonstrate the activity of FLT3 inhibitors, including sorafenib, in targeting the underlying mutation. PROCEDURE: We conducted a retrospective study of 15 pediatric patients with FLT3/ITD+ AML treated with sorafenib within 18 months after receiving HSCT. Sorafenib was administered either as prophylaxis in patients considered at very high risk for relapse (n = 6) or at the time of disease recurrence (n = 9). RESULTS: Sorafenib was initiated at a median of 100 days post HSCT. Overall, 11/15 (73%) of patients experienced medically significant toxicities. Among patients who experienced toxicity, 6/11 (55%) received treatment at doses above what was later determined to be the maximum tolerated dose of sorafenib for pediatric leukemia. Importantly, sorafenib did not appear to exacerbate graft versus host disease. Our findings suggest that sorafenib may be of particular efficacy in patients with minimal residual disease (MRD); all patients who received sorafenib for MRD immediately prior to transplant or with emergence post-HSCT are alive and remain in complete remission at a median of 48 months post HSCT. CONCLUSIONS: Our case series suggests that sorafenib administration is feasible and tolerable in pediatric FLT3/ITD+ AML patients early post HSCT. Ongoing prospective controlled studies are needed to further define the dosing of sorafenib in the post-HSCT period and to determine the optimal context for this treatment approach.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Tandem Repeat Sequences , fms-Like Tyrosine Kinase 3/genetics , Adolescent , Adult , Child , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Neoplasm, Residual , Niacinamide/adverse effects , Niacinamide/therapeutic use , Phenylurea Compounds/adverse effects , Retrospective Studies , Sorafenib
11.
Pediatr Blood Cancer ; 62(2): 335-340, 2015 02.
Article in English | MEDLINE | ID: mdl-25307425

ABSTRACT

BACKGROUND: Humanism and professionalism are virtues intrinsic to the practice of medicine, for which we lack a standard, evidence-based approach for teaching and evaluation. Pediatric hematology-oncology (PHO) fellowship training brings new and significant stressors, making it an attractive setting for innovation in humanism and professionalism training. PROCEDURE: We electronically surveyed a national sample of PHO fellows to identify fellows' educational needs in humanism and professionalism. Next, we developed a case-based, faculty-facilitated discussion curriculum to teach this content within pilot fellowship programs. We assessed whether fellowships would decide to offer the curriculum, feasibility of administering the curriculum, and satisfaction of fellow and faculty participants. RESULTS: Surveys were completed by 187 fellows (35%). A minority (29%) reported that their training program offers a formal curriculum in humanism and/or professionalism. A majority desires more formal teaching on balancing clinical practice and research (85%), coping with death/dying (85%), bereavement (78%), balancing work and personal life (75%), navigating challenging relationships with patients (74%), and depression/burn out (71%). These six topics were condensed into four case-based modules, which proved feasible to deliver at all pilot sites. Ten fellowship programs agreed to administer the novel curriculum. The majority (90%) of responding fellows and faculty reported the sessions touched on issues important for training, stimulated reflective communication, and were valuable. CONCLUSIONS: Pediatric hematology-oncology fellows identify numerous gaps in their training related to humanism and professionalism. This curriculum offers an opportunity to systematically address these educational needs and can serve as a model for wider implementation. Pediatr Blood Cancer 2015;62:335-340. © 2014 Wiley Periodicals, Inc.


Subject(s)
Education, Medical, Graduate/methods , Hematology/education , Humanism , Medical Oncology/education , Professionalism/education , Adult , Attitude of Health Personnel , Curriculum , Female , Humans , Male , Surveys and Questionnaires
12.
J Natl Cancer Inst ; 104(16): 1240-50, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22835387

ABSTRACT

BACKGROUND: Basal cell carcinoma (BCC) is the most common malignancy in the United States. Ionizing radiation is an established risk factor in certain populations, including cancer survivors. We quantified the association between ionizing radiation dose and the risk of BCC in childhood cancer survivors. METHODS: Participants in the Childhood Cancer Survivor Study who reported a BCC (case subjects, n = 199) were matched on age and length of follow-up to three study participants who had not developed a BCC (control subjects, n = 597). The radiation-absorbed dose (in Gy) to the BCC location was calculated based on individual radiotherapy records using a custom-designed dosimetry program. Conditional logistic regression was used to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for associations between demographic and treatment factors, therapeutic radiation dose, and surrogate markers of sun sensitivity (skin and hair color) and the risk of BCC. A linear dose-response model was fitted to evaluate the excess odds ratio per Gy of radiation dose. RESULTS: Among case subjects, 83% developed BCC between the ages of 20 and 39 years. Radiation therapy, either alone or in combination with chemotherapy, was associated with an increased risk of BCC compared with no chemotherapy or radiation. The odds ratio for subjects who received 35 Gy or more to the skin site vs no radiation therapy was 39.8 (95% CI = 8.6 to 185). Results were consistent with a linear dose-response relationship, with an excess odds ratio per Gy of 1.09 (95% CI = 0.49 to 2.64). No other treatment variables were statistically significantly associated with an increased risk of BCC. CONCLUSIONS: Radiation doses to the skin of more than 1 Gy are associated with an increased risk of BCC.


Subject(s)
Carcinoma, Basal Cell/epidemiology , Carcinoma, Basal Cell/etiology , Neoplasms, Radiation-Induced/epidemiology , Neoplasms, Radiation-Induced/etiology , Radiation, Ionizing , Skin Neoplasms/epidemiology , Skin Neoplasms/etiology , Sunlight/adverse effects , Adolescent , Child , Child, Preschool , Cohort Studies , Dose-Response Relationship, Radiation , Female , Hair Color , Humans , Linear Models , Logistic Models , Male , Odds Ratio , Radiotherapy Dosage , Retrospective Studies , Risk Assessment , Risk Factors , Skin/radiation effects , Skin Pigmentation , Survivors/statistics & numerical data , United States/epidemiology
13.
Pediatr Blood Cancer ; 59(4): 756-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22052552

ABSTRACT

The prognosis for children with acute myelogenous leukemia (AML) has improved with overall survival rates of up to 65% [Pui et al. J Clin Oncol 2011; 29: 551-565]. However, the cure rate for AML lags behind that of acute lymphoblastic leukemia. Advances in AML leukemogenesis are leading to refined risk stratification. FMS like tyrosine kinase 3 (FLT3) mutations are independently associated with a poor prognosis. Newer kinase inhibitors, including sorafenib, have shown promise in adult studies. We report three pediatric patients with relapsed AML who achieved a sustained remission with sorafenib. Further trials are necessary to understand the role of sorafenib in pediatric AML.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Adolescent , Child , Female , Humans , Male , Niacinamide/analogs & derivatives , Phenylurea Compounds , Recurrence , Remission Induction , Sorafenib
14.
BMC Cancer ; 9: 411, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19939254

ABSTRACT

BACKGROUND: c-Met is a tyrosine kinase receptor for hepatocyte growth factor/scatter factor (HGF/SF), and both c-Met and its ligand are expressed in a variety of tissues. C-Met/HGF/SF signaling is essential for normal embryogenesis, organogenesis, and tissue regeneration. Abnormal c-Met/HGF/SF signaling has been demonstrated in different tumors and linked to aggressive and metastatic tumor phenotypes. In vitro and in vivo studies have demonstrated inhibition of c-Met/HGF/SF signaling by the small-molecule inhibitor PHA665752. This study investigated c-Met and HGF expression in two neuroblastoma (NBL) cell lines and tumor tissue from patients with NBL, as well as the effects of PHA665752 on growth and motility of NBL cell lines. The effect of the tumor suppressor protein PTEN on migration and proliferation of tumor cells treated with PHA665752 was also evaluated. METHODS: Expression of c-Met and HGF in NBL cell lines SH-EP and SH-SY5Y and primary tumor tissue was assessed by immunohistochemistry and quantitative RT-PCR. The effect of PHA665752 on c-Met/HGF signaling involved in NBL cell proliferation and migration was evaluated in c-Met-positive cells and c-Met-transfected cells. The transwell chemotaxis assay and the MTT assay were used to measure migration and proliferation/cell-survival of tumor cells, respectively. The PPAR-gamma agonist rosiglitazone was used to assess the effect of PTEN on PHA665752-induced inhibition of NBL cell proliferation/cell-survival and migration RESULTS: High c-Met expression was detected in SH-EP cells and primary tumors from patients with advanced-stage disease. C-Met/HGF signaling induced both migration and proliferation of SH-EP cells. Migration and proliferation/cell-survival were inhibited by PHA665752 in a dose-dependent manner. We also found that induced overexpression of PTEN following treatment with rosiglitazone significantly enhanced the inhibitory effect of PHA665752 on NBL-cell migration and proliferation. CONCLUSION: c-Met is highly expressed in most tumors from patients with advanced-stage, metastatic NBL. Furthermore, using the NBL cell line SH-EP as a model, PHA665752 was shown to inhibit cMet/HGF/SF signaling in vitro, suggesting c-Met inhibitors may have efficacy for blocking local progression and/or metastatic spread of c-Met-positive NBL in vivo. These are novel findings for this disease and suggest that further studies of agents targeting the c-Met/HGF axis in NBL are warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Hepatocyte Growth Factor/metabolism , Indoles/pharmacology , Neuroblastoma/metabolism , Proto-Oncogene Proteins c-met/metabolism , Sulfones/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Hypoglycemic Agents/pharmacology , MAP Kinase Signaling System/drug effects , Neoplasm Staging , Neuroblastoma/pathology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-met/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Thiazolidinediones/pharmacology , Transfection
15.
Pediatr Blood Cancer ; 53(1): 90-1, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19301397

ABSTRACT

Atypical hemolytic uremic syndrome remains a challenge to diagnose and treat, with significant acute morbidity and risk for progression to end stage renal disease. Treatment strategies center on plasma exchange but do not necessarily affect the progression of disease. We report the case of a patient with atypical HUS resulting from a mutation in the complement pathway who responded to treatment with steroids and IVIG, therefore avoiding transfusion or plasma exchange.


Subject(s)
Glucocorticoids/therapeutic use , Hemolytic-Uremic Syndrome/drug therapy , Immunoglobulins, Intravenous/therapeutic use , Methylprednisolone/therapeutic use , Blood Transfusion , Hemolytic-Uremic Syndrome/genetics , Humans , Immunologic Factors/therapeutic use , Infant , Male , Membrane Cofactor Protein/genetics , Mutation , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...