125 results on '"Sofia, Haque"'
Search Results
2. Outcomes of intraventricular 131-I-omburtamab and external beam radiotherapy in patients with recurrent medulloblastoma and ependymoma
- Author
-
Kathryn R. Tringale, Suzanne L. Wolden, Matthias Karajannis, Sofia Haque, Luca Pasquini, Onur Yildirim, Marc Rosenblum, Jamal K. Benhamida, Nai-Kong Cheung, Mark Souweidane, Ellen M. Basu, Neeta Pandit-Taskar, Pat B. Zanzonico, John L. Humm, and Kim Kramer
- Subjects
Cancer Research ,Neurology ,Oncology ,Neurology (clinical) - Abstract
Purpose Intraventricular compartmental radioimmunotherapy (cRIT) with 131-I-omburtamab is a potential therapy for recurrent primary brain tumors that can seed the thecal space. These patients often previously received external beam radiotherapy (EBRT) to a portion or full craniospinal axis (CSI) as part of upfront therapy. Little is known regarding outcomes after re-irradiation as part of multimodality therapy including cRIT. This study evaluates predictors of response, patterns of failure, and radiologic events after cRIT. Methods Patients with recurrent medulloblastoma or ependymoma who received 131-I-omburtamab on a prospective clinical trial were included. Extent of disease at cRIT initiation (no evidence of disease [NED] vs measurable disease [MD]) was assessed as associated with progression-free (PFS) and overall survival (OS) by Kaplan–Meier analysis. Results All 27 patients (20 medulloblastoma, 7 ependymoma) had EBRT preceding cRIT: most (22, 81%) included CSI (median dose 2340 cGy, boost to 5400 cGy). Twelve (44%) also received EBRT at relapse as bridging to cRIT. There were no cases of radionecrosis. At cRIT initiation, 11 (55%) medulloblastoma and 3 (43%) ependymoma patients were NED, associated with improved PFS (p = 0.002) and OS (p = 0.048) in medulloblastoma. Most relapses were multifocal. With medium follow-up of 3.0 years (95% confidence interval, 1.8–7.4), 6 patients remain alive with NED. Conclusion For patients with medulloblastoma, remission at time of cRIT was associated with significantly improved survival outcomes. Relapses are often multifocal, particularly in the setting of measurable disease at cRIT initiation. EBRT is a promising tool to achieve NED status at cRIT initiation, with no cases of radiation necrosis.
- Published
- 2023
3. A phase I trial of ganetespib in combination with paclitaxel and trastuzumab in patients with human epidermal growth factor receptor-2 (HER2)-positive metastatic breast cancer
- Author
-
Komal Jhaveri, Rui Wang, Eleonora Teplinsky, Sarat Chandarlapaty, David Solit, Karen Cadoo, James Speyer, Gabriella D’Andrea, Sylvia Adams, Sujata Patil, Sofia Haque, Tara O’Neill, Kent Friedman, Francisco J. Esteva, Clifford Hudis, and Shanu Modi
- Subjects
Ganetespib ,Paclitaxel ,Trastuzumab ,HSP90 inhibitor ,HER2 ,Metastatic breast cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Targeted therapies in HER2-positive metastatic breast cancer significantly improve outcomes but efficacy is limited by therapeutic resistance. HER2 is an acutely sensitive Heat Shock Protein 90 (HSP90) client and HSP90 inhibition can overcome trastuzumab resistance. Preclinical data suggest that HSP90 inhibition is synergistic with taxanes with the potential for significant clinical activity. We therefore tested ganetespib, a HSP90 inhibitor, in combination with paclitaxel and trastuzumab in patients with trastuzumab-refractory HER2-positive metastatic breast cancer. Methods In this phase I dose-escalation study, patients with trastuzumab-resistant HER2-positive metastatic breast cancer received weekly trastuzumab (2 mg/kg) and paclitaxel (80 mg/m2) on days 1, 8, 15, and 22 of a 28-day cycle with escalating doses of ganetespib (100 mg/m2, 150 mg/m2, and a third cohort of 125 mg/m2 if needed) on days 1, 8, and 15. Therapy was continued until disease progression or toxicity. The primary objective was to establish the safety and maximum tolerated dose and/or recommended phase II dose (RP2D) of this therapy. The secondary objectives included evaluation of the effects of ganetespib on the pharmacokinetics of paclitaxel, and to make a preliminary assessment of the efficacy of the combination therapy. Results Dose escalation was completed for the two main cohorts without any observed dose-limiting toxicities. Nine patients received treatment. The median prior lines of anti-HER2 therapy numbered three (range 2–4), including prior pertuzumab in 9/9 patients and ado-trastuzumab emtansine (T-DM1) in 8/9 patients. The most common grade 1/2 adverse events (AEs) were diarrhea, fatigue, anemia, and rash. There were no grade 4 AEs related to ganetespib. The overall response rate was 22% (2/9 patients had partial response) and stable disease was seen in 56% (5/9 patients). The clinical benefit rate was 44% (4/9 patients). The median progression-free survival was 20 weeks (range 8–55). Conclusion The RP2D of ganetespib is 150 mg/m2 in combination with weekly paclitaxel plus trastuzumab. The combination was safe and well tolerated. Despite prior taxanes, pertuzumab, and T-DM1, clinical activity of this triplet regimen in this heavily pretreated cohort is promising and warrants further study in HER2-positive metastatic breast cancer. Trial registration ClinicalTrials.gov NCT02060253 . Registered 30 January 2014.
- Published
- 2017
- Full Text
- View/download PDF
4. The use of single-timepoint images to link administered radioiodine activity (MBq) to a prescribed lesion radiation-absorbed dose (cGy): a regression-based prediction interval tool for the management of well-differentiated thyroid cancer patients
- Author
-
Audrey Mauguen, Ravinder K. Grewal, Finn Augensen, Murad Abusamra, Sonia Mahajan, Vetri Sudar Jayaprakasam, Joseph Osborne, Sofia Haque, Bernadette Z. Y. Wong, Ronald A. Ghossein, James Fagin, Heiko Schӧder, R. Michael Tuttle, Alan Ho, John L. Humm, and Steven M. Larson
- Subjects
Radiology, Nuclear Medicine and imaging ,General Medicine - Abstract
Purpose To introduce a biomarker-based dosimetry method for the rational selection of a treatment activity for patients undergoing radioactive iodine 131I therapy (RAI) for metastatic differentiated thyroid cancer (mDTC) based on single-timepoint imaging of individual lesion uptake by 124I PET. Methods Patients referred for RAI therapy of mDTC were enrolled in institutionally approved protocols. A total of 208 mDTC lesions (in 21 patients) with SUVmax > 1 underwent quantitative PET scans at 24, 48, 72, and 120 h post-administration of 222 MBq of theranostic NaI-124I to determine the individual lesion radiation-absorbed dose. Using a general estimating equation, a prediction curve for biomarker development was generated in the form of a best-fit regression line and 95% prediction interval, correlating individual predicted lesion radiation dose metrics, with candidate biomarkers (“predictors”) such as SUVmax and activity in microcurie per gram, from a single imaging timepoint. Results In the 169 lesions (in 15 patients) that received 131I therapy, individual lesion cGy varied over 3 logs with a median of 22,000 cGy, confirming wide heterogeneity of lesion radiation dose. Initial findings from the prediction curve on all 208 lesions confirmed that a 48-h SUVmax was the best predictor of lesion radiation dose and permitted calculation of the 131I activity required to achieve a lesional threshold radiation dose (2000 cGy) within defined confidence intervals. Conclusions Based on MIRD lesion-absorbed dose estimates and regression statistics, we report on the feasibility of a new single-timepoint 124I-PET-based dosimetry biomarker for RAI in patients with mDTC. The approach provides clinicians with a tool to select personalized (precision) therapeutic administration of radioactivity (MBq) to achieve a desired target lesion-absorbed dose (cGy) for selected index lesions based on a single 48-h measurement 124I-PET image, provided the selected activity does not exceed the maximum tolerated activity (MTA) of Trial registration NCT04462471, Registered July 8, 2020. NCT03647358, Registered Aug 27, 2018.
- Published
- 2023
5. Supplementary Material from A Secondary Mutation in BRAF Confers Resistance to RAF Inhibition in a BRAFV600E-Mutant Brain Tumor
- Author
-
Christine A. Pratilas, Barry S. Taylor, Neal Rosen, David J. Pisapia, Marc K. Rosenblum, Sofia Haque, Katia Manova, Mary Petriccione, Ira J. Dunkel, Sharmeen Uddin, Alice Can Ran Qin, Amy N. Allen, Philip Jonsson, Zhan Yao, and Jiawan Wang
- Abstract
Supplementary Figure S1: Representative H&E, phospho-ERK and FISH images of pre-dabrafenib (3) and post-dabrafenib (4) tumors. Supplementary Figure S2: WES analysis of copy number variation in pre- and postdabrafenib tumors. Supplementary Figure S3: Whole copy number profiles from WES of pre- and posttreatment tumors. Supplementary Figure S4: Homology alignment of BRAF p. L514 with residues in other tyrosine kinases. Supplementary Figure S5: Relative frequency of BRAF variant alleles in SK-BT-DR cells determined by individual clone sequencing. Supplementary Figure S6: BRAF V600E/L514V reduces dabrafenib sensitivity in NIH- 3T3 cells, related to Figure 2. Supplementary Figure S7: BRAF V600E/L514V confers biochemical resistance to dabrafenib over a time course, related to Figure 2G. Supplementary Figure S8: Comparison of IC50, IC75 and IC90 of dabrafenib against A375 cells expressing BRAF V600E and BRAF V600E/L514V, related to Figure 2H. Supplementary Figure S9: The BRAF V600E/L514V double mutant promotes homodimerization, related to Figure 3A and B. Supplementary Figure S10: BRAF L514V alone is hypoactive and associated with decreased ERK signaling that is not sensitive to dabrafenib. Supplementary Figure S11: BRAF V600E/L514V is inhibited by dabrafenib in a purified kinase assay, indicating that it is not a gatekeeper mutation. Supplementary Figure S12: Quantitation of p-MEK and p-ERK immunoblots, related to Figure 4B. Supplementary Figure S13: Comparison of IC50, IC75 and IC90 of trametinib against A375 expressing BRAF V600E and BRAF V600E/L514V, related to Figure 4C. Supplementary Figure S14: The BRAF V600E/L514V mutant mediates resistance to dabrafenib that cannot be completely overcome by trametinib or dabrafenib plus trametinib, related to Figure 4D. Supplementary Figure S15: V5, p-MEK, total MEK immunoblots, and quantitation of p-ERK immunoblots, related to Figure 5A. Supplementary Figure S16: Novel RAF dimer inhibitors, MEK inhibitor and ERK inhibitor equipotently inhibit cell growth in BRAF V600E and V600E/L514V expressing cells. Supplementary Figure S17: Novel RAF dimer inhibitor, MEK inhibitor and ERK inhibitor equipotently inhibit ERK signaling in BRAF V600E and V600E/L514V expressing cells. Supplementary Figure S18: BGB3245 binds mutant BRAF V600E monomer and second site of V600E/L514V dimer with similar affinity. Supplementary Table S1: Mutations identified by WES of pre-dabrafenib and postdabrafenib tumors. Supplementary Table S2: Secondary mutations associated with acquired resistance and occurring in residues homologous with L514 in BRAF. Supplementary Table S3: BRAF L514V allele frequency determined by ddPCR.
- Published
- 2023
6. Enhancing Radioiodine Incorporation in
- Author
-
Vatche, Tchekmedyian, Lara, Dunn, Eric, Sherman, Shrujal S, Baxi, Ravinder K, Grewal, Steven M, Larson, Keith S, Pentlow, Sofia, Haque, R Michael, Tuttle, Mona M, Sabra, Stephanie, Fish, Laura, Boucai, Jamie, Walters, Ronald A, Ghossein, Venkatraman E, Seshan, Jeffrey A, Knauf, David G, Pfister, James A, Fagin, and Alan L, Ho
- Subjects
Iodine Radioisotopes ,Proto-Oncogene Proteins B-raf ,Thyroid Radiology and Nuclear Medicine ,Vemurafenib ,Positron Emission Tomography Computed Tomography ,Mutation ,Antibodies, Monoclonal ,Humans ,Antineoplastic Agents ,Thyroid Neoplasms - Abstract
BACKGROUND: Oncogenic activation of mitogen-activated protein kinase (MAPK) signaling is associated with radioiodine refractory (RAIR) thyroid cancer. Preclinical models suggest that activation of the receptor tyrosine kinase erbB-3 (HER3) mitigates the MAPK pathway inhibition achieved by BRAF inhibitors in BRAF(V600E) mutant thyroid cancers. We hypothesized that combined inhibition of BRAF and HER3 using vemurafenib and the human monoclonal antibody CDX-3379, respectively, would potently inhibit MAPK activation and restore radioactive iodine (RAI) avidity in patients with BRAF-mutant RAIR thyroid cancer. METHODS: Patients with BRAF(V600E) RAIR thyroid cancer were evaluated by thyrogen-stimulated iodine-124 ((124)I) positron emission tomography–computed tomography (PET/CT) at baseline and after 5 weeks of treatment with oral vemurafenib 960 mg twice daily alone for 1 week, followed by vemurafenib in combination with 1000 mg of intravenous CDX-3379 every 2 weeks. Patients with adequate (124)I uptake on the second PET/CT then received therapeutic radioactive iodine ((131)I) with vemurafenb+CDX-3379. All therapy was discontinued two days later. Treatment response was monitored by serum thyroglobulin measurements and imaging. The primary endpoints were safety and tolerability of vemurafenib+CDX-3379, as well as the proportion of patients after vemurafenb+CDX-3379 therapy with enhanced RAI incorporation warranting therapeutic (131)I. RESULTS: Seven patients were enrolled; six were evaluable for the primary endpoints. No grade 3 or 4 toxicities related to CDX-3379 were observed. Five patients had increased RAI uptake after treatment; in 4 patients this increased uptake warranted therapeutic (131)I. At 6 months, 2 patients achieved partial response after (131)I and 2 progression of disease. Next-generation sequencing of 5 patients showed that all had co-occurring telomerase reverse transcriptase promoter alterations. A deleterious mutation in the SWItch/Sucrose Non-Fermentable (SWI/SNF) gene ARID2 was discovered in the patient without enhanced RAI avidity after therapy and an RAI-resistant tumor from another patient that was sampled off-study. CONCLUSIONS: The endpoints for success were met, providing preliminary evidence of vemurafenib+CDX-3379 safety and efficacy for enhancing RAI uptake. Preclinical data and genomic profiling in this small cohort suggest SWI/SNF gene mutations should be investigated as potential markers of resistance to redifferentiation strategies. Further evaluation of vemurafenib+CDX-3379 as a redifferentiation therapy in a larger trial is warranted (ClinicalTrials.gov: NCT02456701).
- Published
- 2023
7. Enhancing Radioiodine Incorporation in BRAF-Mutant, Radioiodine-Refractory Thyroid Cancers with Vemurafenib and the Anti-ErbB3 Monoclonal Antibody CDX-3379: Results of a Pilot Clinical Trial
- Author
-
Vatche Tchekmedyian, Lara Dunn, Eric Sherman, Shrujal S. Baxi, Ravinder K. Grewal, Steven M. Larson, Keith S. Pentlow, Sofia Haque, R. Michael Tuttle, Mona M. Sabra, Stephanie Fish, Laura Boucai, Jamie Walters, Ronald A. Ghossein, Venkatraman E. Seshan, Jeffrey A. Knauf, David G. Pfister, James A. Fagin, and Alan L. Ho
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism - Published
- 2022
8. Toward precision biomarker RAI therapy of well-differentiated thyroid cancer: linking administered radioactivity (MBq) to a prescribed lesion radiation-absorbed dose (cGy)
- Author
-
Audrey Mauguen, Ravinder Grewal, Finn Augensen, Murad Abusamra, Sonia Mahajan, Vetri Sudar Jayaprakasam, Joseph Osborne, Keith Pentlow, Sofia Haque, James Fagin, Heiko Schoder, Michael Tuttle, Alan Ho, John Humm, and Steven Larson
- Abstract
Purpose To introduce a biomarker-based dosimetry method for radioactive iodine 131I therapy (RAI) of metastatic differentiated thyroid cancer (mDTC), adapted to the underlying heterogeneity of lesions’ radiation-absorbed dose (RAD) in cGy, and permitting 1) estimates of RAD/lesion with known precision and 2) optimization of patient-specific administered amount of radioactive iodine (131I) in MBq. Methods Patients referred for RAI therapy of mDTC were enrolled in institutionally approved protocols. A total of 208 mDTC lesions (in 21 patients) with SUV > 1 underwent quantitative PET scans at 24, 48, 72, and 120 hours post-administration of 222 MBq of theranostic NaI-124I to determine the individual lesion RAD (in cGy) based on MIRD 2020 guidance. Using a general estimating equation, a prediction curve for biomarker development was generated in the form of a best-fit regression line and 95% prediction interval, correlating individual predicted lesion RAD metrics, with candidate biomarkers (“predictors”) such as SUV and activity in microcurie per gram, from a single imaging timepoint. Results In the 169 lesions (in 15 patients) that received 131I therapy, individual lesion cGy varied over 3 logs with a median of 22,000 cGy, confirming that heterogeneity of lesion RAD was profound. Initial findings from the prediction curve on all 208 lesions confirmed that 48h SUV was the best predictor of lesion RAD and permitted calculation of the 131I required (in MBq) to achieve a lesional threshold dose for response (2,000 cGy) in more than 95% of lesions. Conclusions Based on MIRD lesion RAD estimates and regression statistics, we propose and report initial feasibility for an 124I-PET-based dosimetry biomarker for RAI in patients with mDTC. This approach enables clinicians to select personalized (precision) therapeutic administration of radioactivity (MBq) to achieve a desired target lesion-absorbed doses (cGy) for > 95% of all lesions using a single 48-hour measurement 124I-PET image. NCT04462471, Registered July 8, 2020. NCT03647358, Registered Aug 27, 2018.
- Published
- 2022
9. Neurocutaneous melanocytosis‐associated malignant melanoma presenting with peritoneal seeding
- Author
-
Ugur Sener, Kevin Elmore, Marc K. Rosenblum, Joanne Porter, Keerthana Jayaseelan, Ashfaq A. Marghoob, Sofia Haque, and Yasmin Khakoo
- Subjects
Pathology ,medicine.medical_specialty ,business.industry ,Melanoma ,Leptomeninges ,Dermatology ,medicine.disease ,Article ,Hydrocephalus ,Meningeal carcinomatosis ,Pediatrics, Perinatology and Child Health ,Cerebrospinal fluid diversion ,Ascites ,medicine ,medicine.symptom ,business ,Complication ,Intracranial pressure - Abstract
Neurocutaneous melanocytosis (NCM) is characterized by melanocyte deposition in the leptomeninges and brain parenchyma, primarily occurring in children with large or giant congenital melanocytic nevi (LCMN) or multiple congenital melanocytic nevi. Patients with NCM may develop hydrocephalus and increased intracranial pressure, which can be managed with ventriculoperitoneal (VP) shunting. We present the case of a 16-month-old girl who developed peritoneal carcinomatosis and malignant ascites following VP shunting for hydrocephalus secondary to NCM to increase awareness of this rare, but serious, complication of cerebrospinal fluid diversion.
- Published
- 2021
10. A phase I study of single-agent perifosine for recurrent or refractory pediatric CNS and solid tumors.
- Author
-
Oren J Becher, Nathan E Millard, Shakeel Modak, Brian H Kushner, Sofia Haque, Ivan Spasojevic, Tanya M Trippett, Stephen W Gilheeney, Yasmin Khakoo, David C Lyden, Kevin C De Braganca, Jill M Kolesar, Jason T Huse, Kim Kramer, Nai-Kong V Cheung, and Ira J Dunkel
- Subjects
Medicine ,Science - Abstract
The PI3K/Akt/mTOR signaling pathway is aberrantly activated in various pediatric tumors. We conducted a phase I study of the Akt inhibitor perifosine in patients with recurrent/refractory pediatric CNS and solid tumors. This was a standard 3+3 open-label dose-escalation study to assess pharmacokinetics, describe toxicities, and identify the MTD for single-agent perifosine. Five dose levels were investigated, ranging from 25 to 125 mg/m2/day for 28 days per cycle. Twenty-three patients (median age 10 years, range 4-18 years) with CNS tumors (DIPG [n = 3], high-grade glioma [n = 5], medulloblastoma [n = 2], ependymoma [n = 3]), neuroblastoma (n = 8), Wilms tumor (n = 1), and Ewing sarcoma (n = 1) were treated. Only one DLT occurred (grade 4 hyperuricemia at dose level 4). The most common grade 3 or 4 toxicity at least possibly related to perifosine was neutropenia (8.7%), with the remaining grade 3 or 4 toxicities (fatigue, hyperglycemia, fever, hyperuricemia, and catheter-related infection) occurring in one patient each. Pharmacokinetics was dose-saturable at doses above 50 mg/m2/day with significant inter-patient variability, consistent with findings reported in adult studies. One patient with DIPG (dose level 5) and 4 of 5 patients with high-grade glioma (dose levels 2 and 3) experienced stable disease for two months. Five subjects with neuroblastoma (dose levels 1 through 4) achieved stable disease which was prolonged (≥11 months) in three. No objective responses were noted. In conclusion, the use of perifosine was safe and feasible in patients with recurrent/refractory pediatric CNS and solid tumors. An MTD was not defined by the 5 dose levels investigated. Our RP2D is 50 mg/m2/day.
- Published
- 2017
- Full Text
- View/download PDF
11. 213 A Positive Impact on Patterns of Relapse in Diffuse Intrinsic Pontine Glioma after Convection-Enhanced Delivery of Omburtamab
- Author
-
Evan D. Bander, Andrew L.A. Garton, Luca Pasquini, Onur Yildirim, Ahmet Ilica, Morgan E. Freret, Maria Donzelli, Sofia Haque, and Mark M. Souweidane
- Subjects
Surgery ,Neurology (clinical) - Published
- 2023
12. A Potential Role For Apparent Diffusion Coefficient in the Diagnosis of Trilateral Retinoblastoma
- Author
-
Jasmine H. Francis, Brian P. Marr, David H. Abramson, Sasan Karimi, Sameer Farouk Sait, Mark M. Souweidane, Ira J. Dunkel, Karim J Rebeiz, and Sofia Haque
- Subjects
Male ,medicine.medical_specialty ,Trilateral retinoblastoma ,Retinal Neoplasms ,Neuroimaging ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pineal Cyst ,Humans ,Medicine ,Effective diffusion coefficient ,Brain magnetic resonance imaging ,Bilateral retinoblastoma ,Retrospective Studies ,Extramural ,business.industry ,Retinoblastoma ,Infant ,Hematology ,medicine.disease ,body regions ,Diffusion Magnetic Resonance Imaging ,Oncology ,Child, Preschool ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Female ,Radiology ,business ,030215 immunology - Abstract
We attempted to investigate the potential role for apparent diffusion coefficient (ADC) to diagnose trilateral retinoblastoma (TRb) by retrospectively reviewing brain magnetic resonance images of retinoblastoma patients. Observations: The median ADC measured 620.95 for TRb (n=6) and 1238.5 for normal pineal gland in bilateral retinoblastoma (n=8). Monitoring ADC trends aided in establishing the appropriate diagnoses in 3 patients (2 TRb, 1 benign pineal cyst). Conclusions: Our results provide baseline reference data and describe the importance of downward trending ADC which should prompt consideration of TRb. Unchanged high/nonrestricted values (>1000) may distinguish those with benign pineal tissue and obviate invasive neurosurgical procedures.
- Published
- 2020
13. A Phase 1b Study of Cetuximab and BYL719 (Alpelisib) Concurrent with Intensity Modulated Radiation Therapy in Stage III-IVB Head and Neck Squamous Cell Carcinoma
- Author
-
Nora Katabi, Matthew G. Fury, Lara Dunn, Shrujal S. Baxi, Loren S. Michel, Sofia Haque, Richard J. Wong, Nadeem Riaz, Sean McBride, David G. Pfister, Han Xiao, Alan L. Ho, Eric J. Sherman, and Nancy Y. Lee
- Subjects
Adult ,Male ,Oncology ,Radiation-Sensitizing Agents ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Class I Phosphatidylinositol 3-Kinases ,medicine.medical_treatment ,Cetuximab ,Loading dose ,Article ,030218 nuclear medicine & medical imaging ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Mucositis ,Humans ,Radiology, Nuclear Medicine and imaging ,Adverse effect ,neoplasms ,Aged ,Phosphoinositide-3 Kinase Inhibitors ,Radiation ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,TOR Serine-Threonine Kinases ,Cancer ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Head and neck squamous-cell carcinoma ,Radiation therapy ,Thiazoles ,stomatognathic diseases ,Regimen ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Mutation ,Radiotherapy, Intensity-Modulated ,business ,medicine.drug - Abstract
Purpose Activation of the PI3K/mTOR signaling pathway is common in head and neck squamous cell carcinoma (HNSCC). BYL719 is an α-specific PI3K inhibitor that is synergistic and efficacious when combined with cetuximab, a Food and Drug Administration–approved radiosensitizing agent in the treatment of HNSCC. The agent independently has been shown to enhance radiosensitivity. This study evaluates the addition of BYL719 to cetuximab and radiation in the treatment of locally advanced HNSCC. Methods and Materials This is a single-institution, phase 1 study. Patients with American Joint Committee on Cancer seventh edition stage III to IVB HNSCC received standard cetuximab (400 mg/m2 intravenous loading dose) before intensity modulated radiation therapy (IMRT) followed by 250 mg/m2 weekly infusions during IMRT. BYL719 was given orally during IMRT in 3 dose levels: (1) 200 mg/d, (2) 250 mg/d, or (3) 300 mg/d in a standard 3 + 3 dose-escalation design. Results Eleven patients were evaluable. Dose level 2 was the maximum tolerated dose for BYL719. Two patients on dose level 3 had dose-limiting toxicities of oral mucositis that required a dose reduction of BYL719. One patient on dose level 2 had a dose-limiting toxicity of nausea that led to withdrawal of on-study treatment. Related grade 3 or higher adverse events consisted of decreased lymphocyte count, oral mucositis, dysphagia, hyperglycemia, maculopapular rash, and palmar-plantar erythrodysesthesia syndrome. All 11 patients had a complete response on posttreatment imaging, and 10 remain disease free. Of the 8 patients with mutational analysis, 1 had an activating PIK3CA mutation associated with a rapid response on serial intratreatment magnetic resonance imaging scans. Conclusions The recommended phase 2 dose of BYL719 is 250 mg/d in combination with cetuximab and IMRT in patients with locally advanced HNSCC. Further evaluation of the addition of BYL719 to the platinum-sparing regimen of cetuximab and IMRT in the treatment of locally advanced HNSCC is warranted.
- Published
- 2020
14. Magnetic Resonance Imaging Screening for Trilateral Retinoblastoma
- Author
-
Ira J. Dunkel, Danielle Novetsky Friedman, Sofia Haque, Sana Qureshi, Mark M. Souweidane, David H. Abramson, and Jasmine H. Francis
- Subjects
0303 health sciences ,medicine.medical_specialty ,medicine.diagnostic_test ,Trilateral retinoblastoma ,Retinoblastoma ,business.industry ,Incidence (epidemiology) ,Cancer ,Magnetic resonance imaging ,Gene mutation ,medicine.disease ,03 medical and health sciences ,Ophthalmology ,0302 clinical medicine ,Hereditary Retinoblastoma ,Cohort ,030221 ophthalmology & optometry ,medicine ,Radiology ,business ,030304 developmental biology - Abstract
Purpose Magnetic resonance imaging (MRI) has been used for baseline brain imaging and afterward as a screening tool for trilateral retinoblastoma (TRB), but there is no consensus on timing or frequency of screening worldwide. In this study, a cohort of hereditary retinoblastoma patients at increased risk for TRB was identified and the usefulness of aggressive neuroimaging was examined. Design Retrospective review of the medical records and MRI reports of patients with retinoblastoma treated at Memorial Sloan Kettering Cancer Center between January 1, 2006, and December 31, 2016. Participants Three hundred forty-nine total patients with retinoblastoma, including 215 hereditary retinoblastoma patients in the screening group. Methods We reviewed 804 MRI studies of the orbit or brain. Patient and disease characteristics, including laterality, family history, and gene mutation status were analyzed. The impression of every MRI was coded 1 to 5, each value representing a different abnormality. Main Outcome Measures We calculated the incidence of TRB in patients with germline disease as well as the incidence of screening MRI scans showing TRB. Results Among our hereditary retinoblastoma screening cohort (n=215) 4 patients with TRB were identified on screening MRI. All 4 patients showed bilateral disease, pineal gland tumors, and a latency period of at least 1 year. Three of the 4 were deceased by the end of the study. The incidence of TRB diagnosis was 1.9% (95% confidence interval [CI], 0.7%–4.9%). Of the 804 screening MRI scans performed on the screening cohort, 691 (86%) were unremarkable and 4 reported a lesion suspicious for TRB. The overall incidence of detecting TRB on screening MRI in the at-risk cohort was 0.5% (95% CI, 0.2%–1.3%) with a number needed to treat of 202. Conclusions All cases of TRB in our center during the study period developed before the patient was 3 years of age and after a total of only 4 lifetime MRIs. Overall survival from TRB was not improved as a result of screening, and many false-positive results required additional, subsequent MRI scans with anesthesia.
- Published
- 2020
15. Next-generation sequencing of cerebrospinal fluid for clinical molecular diagnostics in pediatric, adolescent and young adult brain tumor patients
- Author
-
Alexandra M Miller, Luca Szalontay, Nancy Bouvier, Katherine Hill, Hamza Ahmad, Johnathan Rafailov, Alex J Lee, M Irene Rodriguez-Sanchez, Onur Yildirim, Arti Patel, Tejus A Bale, Jamal K Benhamida, Ryma Benayed, Maria E Arcila, Maria Donzelli, Ira J Dunkel, Stephen W Gilheeney, Yasmin Khakoo, Kim Kramer, Sameer F Sait, Jeffrey P Greenfield, Mark M Souweidane, Sofia Haque, Audrey Mauguen, Michael F Berger, Ingo K Mellinghoff, and Matthias A Karajannis
- Subjects
Cancer Research ,Young Adult ,Oncology ,Adolescent ,Brain Neoplasms ,Liquid Biopsy ,High-Throughput Nucleotide Sequencing ,Humans ,Neurology (clinical) ,Pathology, Molecular ,Child ,Pediatric Neuro-Oncology - Abstract
Background Safe sampling of central nervous system tumor tissue for diagnostic purposes may be difficult if not impossible, especially in pediatric patients, and an unmet need exists to develop less invasive diagnostic tests. Methods We report our clinical experience with minimally invasive molecular diagnostics using a clinically validated assay for sequencing of cerebrospinal fluid (CSF) cell-free DNA (cfDNA). All CSF samples were collected as part of clinical care, and results reported to both clinicians and patients/families. Results We analyzed 64 CSF samples from 45 pediatric, adolescent and young adult (AYA) patients (pediatric = 25; AYA = 20) with primary and recurrent brain tumors across 12 histopathological subtypes including high-grade glioma (n = 10), medulloblastoma (n = 10), pineoblastoma (n = 5), low-grade glioma (n = 4), diffuse leptomeningeal glioneuronal tumor (DLGNT) (n = 4), retinoblastoma (n = 4), ependymoma (n = 3), and other (n = 5). Somatic alterations were detected in 30/64 samples (46.9%) and in at least one sample per unique patient in 21/45 patients (46.6%). CSF cfDNA positivity was strongly associated with the presence of disseminated disease at the time of collection (81.5% of samples from patients with disseminated disease were positive). No association was seen between CSF cfDNA positivity and the timing of CSF collection during the patient’s disease course. Conclusions We identified three general categories where CSF cfDNA testing provided additional relevant diagnostic, prognostic, and/or therapeutic information, impacting clinical assessment and decision making: (1) diagnosis and/or identification of actionable alterations; (2) monitor response to therapy; and (3) tracking tumor evolution. Our findings support broader implementation of clinical CSF cfDNA testing in this population to improve care.
- Published
- 2022
16. DIPG-53. Long-term survival from a Phase 1 dose-escalation trial using convection-enhanced delivery (CED) of radioimmunotherapeutic124I-omburtamab for treatment of diffuse intrinsic pontine glioma (DIPG)
- Author
-
Mark M Souweidane, Kim Kramer, Neeta Pandit-Tasker, Sofia Haque, Pat Zanzonico, Jorge Carrasquillo, Serge K Lyashchenko, Sunitha B Thakur, Yasmin Khakoo, Ira J Dunkel, Maria Donzelli, Jason S Lewis, Nai-Kong V Cheung, Steve M Larson, Anne S Reiner, Katherine S Panageas, Nicole Manino, and John Rømer Nielsen
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: Median survival from DIPG is less than one year. In a phase 1 dose escalation study (clinicaltrials.gov NCT01502917) 124I-omburtamab targeting B7-H3 was administered intratumorally using CED. METHODS: CED was performed between 4-14 weeks post radiation therapy. Using a 3 + 3 design, 124I-omburtamab was escalated from 0.25-10.0 mCi and infusion volumes (Vi) from 250-10,000 µl with serial 124I PET/CT performed up to ~1 week post-administration. Toxicities were assessed for 30 days. Dose escalation safety was evaluated. Survival was calculated using Kaplan-Meier statistics. RESULTS: 46 children were treated and evaluable for toxicity and survival;4 patients received partial doses and were evaluable for toxicity only. Three patients experienced dose limiting toxicities. Eleven patients had transient treatment related grade 3 toxicities with no grade 4 or 5 toxicities. Grade 3 nervous system toxicities included: muscular weakness(n=8), dysarthria(n=4), ataxia(n=3), dysphagia(n=3), and gait disturbance(n=1). Lesion absorbed doses ranged from 1,000-1,500cGy/mCi, with lesion-to-whole body radiation absorbed-dose ratios of ~900. A dose of 8mCi and infusion volume of 8,000 µl is safe and may provide a distribution volume up to 20cm3. Median survival was 15.3 months (n =46, 95% CI 12.7, 17.3). Survival rate estimates (95% CI) at 1, 2, 3 and 5 years were 0.67 (0.55;0.82); 0.18 (0.09;0.35); 0.10 (0.04;0.26); and 0.05 (0.01;0.20). Four patients survived >3 years; two remain alive (61+ and 106+ months);two have died (44 and 53 month) with distant CNS disease and one with extra-CNS metastasis. CONCLUSION: Administration of escalating doses and volumes of 124I-omburtamab via CED was a viable option for this patient subgroup. The median overall survival was increased 3-4 months compared to historical controls. Anecdotal long-term survival if validated with a planned phase 2 trial would support the concept of whole neuroaxis treatment in combination with CED in a subset of DIPG patients.
- Published
- 2022
17. HGG-06. Phase 2 Study of Veliparib and Local Irradiation, Followed by Maintenance Veliparib and Temozolomide, in Patients with Newly Diagnosed High-Grade Glioma without H3 K27M or BRAF Mutations: A Report from the Children's Oncology Group ACNS1721 Study
- Author
-
Matthias Karajannis, Arzu Onar-Thomas, Patricia Baxter, Nina Butingan, Christine Fuller, Amar Gajjar, Sofia Haque, Nada Jabado, Tong Lin, John Lucas, Shannon MacDonald, Celeste Matsushima, Namrata Patel, Christopher Pierson, Linda Springer, Eileen Stark, Mark Souweidane, Michael Walsh, Wafik Zaky, Maryam Fouladi, and Kenneth Cohen
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND: The outcome for pediatric patients with high-grade glioma (HGG) remains poor. Veliparib, a potent oral PARP1/2 inhibitor, enhances the activity of radiotherapy and DNA-damaging chemotherapy. Preclinical data indicates that veliparib crosses the blood-brain-barrier and enhances the efficacy of radiotherapy and temozolomide in IDH mutant and wild-type HGG models. ACNS1721 was a single-arm, non-randomized phase 2 clinical trial designed to determine whether treatment with veliparib and radiotherapy, followed by the poly (ADP-ribose) polymerase (PARP) inhibitor veliparib and temozolomide, improves progression-free survival (PFS) in pediatric patients with newly diagnosed HGG without H3 K27M or BRAF mutations compared to patient level data from historical cohorts with closely matching clinical and molecular features. METHODS: Following surgical resection, newly diagnosed children with non-metastatic HGG were screened by rapid central pathology review and molecular testing. Eligible patients without somatic H3 K27M or BRAF mutations were enrolled on Stratum 1 (IDH wild-type) or Stratum 2 (IDH mutant). Protocol radiochemotherapy consisted of involved field radiotherapy with concurrent veliparib at 65 mg/m2 twice daily. Adjuvant chemotherapy consisted of up to 10 cycles of veliparib 25 mg/m2 twice daily and temozolomide 135 mg/m2 once daily for 5 days every 4 weeks. RESULTS: Both strata were closed to accrual for futility after planned interim analyses. Among the 23 eligible patients who enrolled on Stratum 1 and received protocol therapy, the 1-year progression-free survival (PFS) was 0.29 (SE = 0.09) and 1-year overall survival (OS) was 0.67 (SE = 0.10). Among the 14 eligible patients who enrolled on Stratum 2 and received protocol therapy, the 1-year PFS was 0.57 (SE = 0.15) and 1-year OS was 0.90 (SE = 0.09). CONCLUSION: Rapid central pathology review and molecular testing was feasible. The protocol therapy was well tolerated but failed to improve outcome compared to clinically and molecularly matched historical control cohorts.
- Published
- 2022
18. CTNI-31. COG ACNS1721: PHASE 2 STUDY OF VELIPARIB AND LOCAL IRRADIATION, FOLLOWED BY MAINTENANCE VELIPARIB AND TEMOZOLOMIDE, IN PATIENTS WITH NEWLY DIAGNOSED HIGH-GRADE GLIOMA WITHOUT H3 K27M OR BRAF MUTATIONS
- Author
-
Matthias Karajannis, Arzu Onar Thomas, Patricia Baxter, Nina Butingan, Christine Fuller, Amar Gajjar, Sofia Haque, Nada Jabado, Tong Lin, John Lucas, Shannon MacDonald, Celeste Matsushima, Namrata Patel, Christopher Pierson, Linda Springer, Eileen Stark, Mark Souweidane, Michael Walsh, Wafik Zaky, Maryam Fouladi, and Kenneth Cohen
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
BACKGROUND The outcome for pediatric patients with high-grade glioma (HGG) remains poor. Veliparib, a potent oral PARP1/2 inhibitor, enhances the activity of radiotherapy and DNA-damaging chemotherapy. Preclinical data indicates that veliparib crosses the blood-brain-barrier and enhances the efficacy of radiotherapy and temozolomide in IDH mutant and wild-type HGG models. ACNS1721 was a single-arm, non-randomized phase 2 clinical trial designed to determine whether treatment with veliparib and radiotherapy, followed by the poly (ADP-ribose) polymerase (PARP) inhibitor veliparib and temozolomide, improves progression-free survival (PFS) in pediatric patients with newly diagnosed HGG without H3 K27M or BRAF mutations compared to patient level data from historical cohorts with closely matching clinical and molecular features. METHODS Following surgical resection, newly diagnosed children with non-metastatic HGG were screened by rapid central pathology review and molecular testing. Eligible patients without somatic H3 K27M or BRAF mutations were enrolled on Stratum 1 (IDH wild-type) or Stratum 2 (IDH mutant). Protocol radiochemotherapy consisted of involved field radiotherapy with concurrent veliparib at 65 mg/m2 twice daily. Adjuvant chemotherapy consisted of up to 10 cycles of veliparib 25 mg/m2 twice daily and temozolomide 135 mg/m2 once daily for 5 days every 4 weeks. RESULTS Both strata were closed to accrual for futility after planned interim analyses. Among the 23 eligible patients who enrolled on Stratum 1 and received protocol therapy, the 1-year progression-free survival (PFS) was 0.29 (SE = 0.09) and 1-year overall survival (OS) was 0.67 (SE = 0.10). Among the 14 eligible patients who enrolled on Stratum 2 and received protocol therapy, the 1-year PFS was 0.57 (SE = 0.15) and 1-year OS was 0.90 (SE = 0.09). CONCLUSION Rapid central pathology review and molecular testing was feasible. The protocol therapy was well tolerated but failed to improve outcome compared to clinically and molecularly matched historical control cohorts.
- Published
- 2022
19. Malignant transformation of a polymorphous low grade neuroepithelial tumor of the young (PLNTY)
- Author
-
Marc Ladanyi, Rupa Juthani, Mary Fowkes, Matthias A. Karajannis, Benjamin Liechty, Jamal Benhamida, Razia B Akhtar, Sameer Farouk Sait, Ryan Ptashkin, Martin Sill, Marc K. Rosenblum, Sofia Haque, Tejus Bale, Justyna Sadowska, and Liliana Villafania
- Subjects
Pathology ,medicine.medical_specialty ,Adolescent ,Brain Neoplasms ,Brain Edema ,Chemoradiotherapy ,Biology ,Magnetic Resonance Imaging ,Neoplasms, Neuroepithelial ,Article ,Pathology and Forensic Medicine ,Malignant transformation ,Neuroepithelial cell ,Cellular and Molecular Neuroscience ,Temozolomide ,medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 3 ,Female ,Neurology (clinical) ,Gene Fusion ,Antineoplastic Agents, Alkylating ,Microtubule-Associated Proteins - Published
- 2020
20. Phase II Study of Lenvatinib in Patients With Progressive, Recurrent or Metastatic Adenoid Cystic Carcinoma
- Author
-
Shrujal S. Baxi, Eric J. Sherman, Cristina R. Antonescu, Crystal Tran, Nora Katabi, Vatche Tchekmedyian, David G. Pfister, Irina Ostrovnaya, Alan L. Ho, Lara Dunn, and Sofia Haque
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Pathology ,medicine.medical_specialty ,Adenoid cystic carcinoma ,Phases of clinical research ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Neoplasm Recurrence ,RAPID COMMUNICATIONS ,Carcinoma ,Humans ,Medicine ,In patient ,Neoplasm Metastasis ,Aged ,Salivary gland ,business.industry ,Phenylurea Compounds ,Disease progression ,Middle Aged ,medicine.disease ,Carcinoma, Adenoid Cystic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Disease Progression ,Quinolines ,Female ,Neoplasm Recurrence, Local ,business ,Lenvatinib - Abstract
PURPOSE Recurrent or metastatic adenoid cystic carcinoma (R/M ACC) is a malignant neoplasm of predominantly salivary gland origin for which effective therapies are lacking. We conducted a phase II trial evaluating the multitargeted tyrosine kinase inhibitor lenvatinib in patients with R/M ACC. PATIENTS AND METHODS This study was conducted with a two-stage minimax design. Patients with histologically confirmed R/M ACC of any primary site with radiographic and/or symptomatic progression were eligible. Any prior therapy was allowed except previous lenvatinib. Patients received lenvatinib 24 mg orally per day. The primary end point was overall response rate. Secondary end points were progression-free survival and safety. An exploratory analysis of how MYB expression and genomic alterations relate to outcomes was conducted. RESULTS Thirty-three patients were enrolled; 32 were evaluable for the primary end point. Five patients (15.6%) had a confirmed partial response, 24 patients (75%) had stable disease, two patients (6.3%) discontinued treatment as a result of toxicity before the first scan, and one patient (3.1%) had progression of disease as best response. Median progression-free survival time was 17.5 months (95% CI, 7.2 months to not reached), although only eight progression events were observed. Patients otherwise were removed for toxicity (n = 5), as a result of withdrawal of consent (n = 9), or at the treating physician’s discretion (n = 6). Twenty-three patients required at least one dose modification, and 18 of 32 patients discontinued lenvatinib for drug-related issues. The most common grade 3 or 4 adverse events were hypertension (n = 9; 28.1%) and oral pain (n = 3; 9.4%). Three grade 4 adverse events were observed (myocardial infarction, n = 1; posterior reversible encephalopathy syndrome, n = 1; and intracranial hemorrhage, n = 1). CONCLUSION This trial met the prespecified overall response rate primary end point, demonstrating antitumor activity with lenvatinib in R/M ACC patients. Toxicity was comparable to previous studies, requiring monitoring and management.
- Published
- 2019
21. A pilot study of neoadjuvant cemiplimab with platinum-doublet chemotherapy and cetuximab in patients with resectable, locally advanced head and neck squamous cell carcinoma (HNSCC)
- Author
-
Lara Dunn, Eric Jeffrey Sherman, Alan Loh Ho, Ian Ganly, Nadeem Riaz, Luc Morris, Kin Wai (Tony) Hung, Anuja Kriplani, Jennifer R. Cracchiolo, Marc Cohen, Jay Boyle, Snehal G. Patel, Sofia Haque, Nora Katabi, Ronald A Ghossein, Sean Matthew McBride, Loren S. Michel, Richard J. Wong, Nancy Y. Lee, and David G. Pfister
- Subjects
Cancer Research ,Oncology - Abstract
TPS6109 Background: Definitive treatment of locally advanced HNSCC can require radical surgery and reconstruction often resulting in unacceptable functional consequences. Radiotherapy, often with concurrent chemotherapy, is administered postoperatively to achieve the best chance for cure. Induction chemotherapy has previously been shown to reduce the extent of surgical resection and need for adjuvant radiation (RT). The purpose of this trial is to evaluate if an induction regimen combining cytotoxic chemotherapy, EGFR targeting, and immune checkpoint blockade can pathologically downstage resectable HNSCC sufficiently to decrease surgical morbidity and justify omission of adjuvant RT-based therapy. Compared to standard docetaxel, cisplatin, and 5-FU (TPF), docetaxel, cisplatin, and cetuximab (TPC) has been shown to be a therapeutic alternative with a more favorable toxicity profile. Targeting PD-1 alone can induce significant pathologic responses in resectable HNSCC patients. Combining PD-1 inhibitors with cetuximab has shown promising activity in incurable HNSCC; cetuximab may optimize the tumor immune microenvionment for PD-1 therapy by stimulating IFN-gamma secretion to increase dendritic cell maturation and CD8 T cell expression of PD1. Based on this rationale, we are evaluating the novel induction regimen of platinum, docetaxel, cetuximab plus cemiplimab (anti-PD1 antibody). Methods: This is a 10-patient pilot study for locally advanced, resectable HNSCC patients for whom standard management requires adjuvant RT +/- chemotherapy. Patients will receive neoadjuvant treatment with a loading dose of cetuximab and cemiplimab followed by 3 cycles of cisplatin or carboplatin, docetaxel, cetuximab and cemiplimab followed by definitive surgical resection of the primary site +/- neck dissection(s). Post-operative RT +/- radiosensitizing agent(s) will be administered per standard of care (SOC) based on pathologic staging (rather than clinical staging at presentation). If the pathologic stage following induction and surgery is ypT0-2N0 without adverse features, adjuvant RT will not be administered and 6 months of adjuvant cemiplimab will be given. Otherwise, patients will receive SOC adjuvant RT-based treatment. The primary endpoint is safety and tolerability. Secondary endpoints include feasibility assessed by the number of patients whose definitive surgery was delayed due to toxicity and quantifying the number in whom clinical to pathologic downstaging is achieved and the planned surgery and/or need for adjuvant-RT based therapy is modified. Exploratory endpoints include evaluating the association between biomarkers in the tumor microenvironment and peripheral blood with pathologic response. 8 of 10 patients have been enrolled. Clinical trial information: NCT04722523.
- Published
- 2022
22. PATH-16. Noninvasive diagnosis of gliomas through CSF cfDNA sequencing in pediatric and adolescent and young adult (AYA) patients
- Author
-
Katherine Hill, Alexandra M Miller, Bryan Kincheon Li, Nancy Bouvier, Shanita Li, Tina Alano, Seyram Doe-Tetteh, Claudia Huereca, Sara DiNapoli, Alex Lee, Luca Szalontay, Cecile Riviere-Cazaux, Terry C Burns, Sofia Haque, Tejus Bale, Jamal Benhamida, Snjezana Dogan, Chad Vanderbilt, Dara Ross, Jason Chang, Maria Donzelli, Ira J Dunkel, Sameer Farouk Sait, Yasmin Khakoo, Stephen Gilheeney, Mark Souweidane, Jeffrey Greenfield, Michael Berger, Ryma Benayed, Maria E Arcila, Marc Ladanyi, Ingo Mellinghoff, and Matthias Karajannis
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
PURPOSE: A subset of pediatric, adolescent and young adult (AYA) gliomas are located in the brainstem, eloquent locations, or present with diffuse/leptomeningeal disease, and are associated with high risk and low yield of biopsy. At the same time, accurate molecular diagnosis is necessary to direct optimal therapy. In such cases, analysis of cell free DNA (cfDNA) form cerebral spinal fluid (CSF) may represent a diagnostic alternative to biopsy. METHODS: We investigated the utility of CSF cfDNA sequencing through a stepwise approach, using clinically validated, targeted molecular assays. Testing was performed using a broad hybrid capture next generation sequencing assay (MSK-IMPACT) and subsequent targeted digital droplet PCR in a subset of cases. RESULTS: We analyzed 17 CSF samples from 17 pediatric (n=6) and AYA (n=11) glioma patients with primary or recurrent disease. Thirteen had tumors located within the brainstem, and four had leptomeningeal involvement. Somatic alterations were detected in 12/17 samples (71%). In 3/4 patients with leptomeningeal involvement, cfDNA testing revealed a BRAF fusion consistent with the diagnosis of diffuse leptomeningeal glioneuronal tumor (DLGNT). Among the 13 patients with brainstem involvement, four had somatic H3 K27M mutations, three had IDH mutations, and one had TP53 and ATRX mutations; five patients had no detectable mutations. CONCLUSION: In our analysis, we found that established glioma hotspot mutations were able to be detected within the CSF. We propose that in patients for whom tissue biopsy is high risk, not feasible, or tissue was nondiagnostic, CSF cfDNA sequencing has a substantial diagnostic yield and should be considered as a valuable novel diagnostic tool. Ongoing research is aiming to further increase the sensitivity of cfDNA testing, especially in patients with very low levels of CSF cfDNA.
- Published
- 2022
23. A pilot study of trametinib in combination with paclitaxel in the treatment of anaplastic thyroid cancer
- Author
-
Eric Jeffrey Sherman, Loren S. Michel, Anuja Kriplani, Lara Dunn, Sofia Haque, Deborah Bang, Sarah Stein, David G. Pfister, and Alan Loh Ho
- Subjects
Cancer Research ,Oncology - Abstract
6088 Background: Anaplastic Thyroid Cancer (ATC) is a rare and highly aggressive tumor with extremely poor prognosis. Outside of the recent approval of dabrafenib/trametinib for BRAF mutant tumors, there are no other standard treatment available for metastatic ATC. The majority of ATC is driven through the MAPK pathway. Data in lung cancer suggested synergy with trametinib, a MEK inhibitor, and taxanes. Methods: In this pilot study we used Trametinib (2 mg) daily with Paclitaxel (80 mg/m2) administered weekly for the first 3 weeks out the 4 week-cycle. Restaging imaging was performed every 6-8 weeks (1.5-2 cycles). Eligible patients had ATC with baseline ECOG performance status ≤ 1 and were enrolled at Memorial Sloan Kettering Cancer Center. Prior treatment allowed. Prior brain metastases were allowed if treated and stable off of steroids. Primary objective was PFS at 6 months with the target of > 2 subjects out of 12 at the time point. Results: 12 patients (6 men and 6 women) were enrolled between 11/2017 and 10/2021. Seven (58%) had prior radiation to the neck; 4 (33%) had prior treatment (not including with radiation) for ATC; 1 (8%) had prior brain metastases. Three (25%) partial responses were reported, and five (41.67%) reported stable disease. Subjects with partial responses had a BRAF V600E mutation (1), BRAF fusion gene (1), and a RAS mutation (1). Median time on treatment was 10.5 weeks (3-47+ weeks). Median overall survival was 26 weeks (3-59+). Six-month progression free survival (PFS) was achieved in 3 patient (25%), one of whom remains on study. 2 patients discontinued treatment due to unacceptable toxicity. The most frequent adverse events observed (all grades) were anemia (75%), increased AST, diarrhea and leukopenia (all 50%). Grade 3/4 AEs included neutropenia (25%), with anemia, AST increased, febrile neutropenia, and lymphopenia, all 16.67%. Grade 4 reactions included lymphopenia (n = 2) and leukopenia (n = 1). Conclusions: Our target progression-free survival (PFS) at 6 months was observed on this study. The combination of trametinib and paclitaxel should be evaluated in a larger cohort of patients in the future. Clinical trial information: NCT03085056.
- Published
- 2022
24. RARE-10. Neurocutaneous melanocytosis-associated hydrocephalus: the MSK experience from 2001-2022
- Author
-
Scarlett Rodriguez, Ugur Sener, Kevin Elmore, Sofia Haque, Stephanie Suser, Jeffrey Greenfield, Maria Donzelli, Courtnee DePass, John Pugh, Joanne Porter, Nathan Meeker, Elizabeth Wells, Ashfaq Marghoob, and Yasmin Khakoo
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
OBJECTIVE: We hypothesize that patients with neurocutaneous melanocytosis-associated melanoma and ventriculoperitoneal shunts are at risk of developing intraperitoneal spread of melanoma. BACKGROUND: Neurocutaneous melanocytosis, a rare condition characterized by excessive proliferation and deposition of melanocytes in the leptomeninges and brain parenchyma, typically occurs in children with large congenital melanocytic nevi and multiple smaller congenital nevi. These patients are at heightened risk for developing NRAS+ melanomas in the central nervous system, which in turn may lead to symptomatic hydrocephalus requiring cerebrospinal fluid diversion for symptom relief. METHODS: Retrospective single-institution study of patients with histologically or radiographically confirmed NCM evaluated at Memorial Sloan Kettering Cancer Center (MSKCC) from 2001-2022. RESULTS: Of the 47 patients with a diagnosis of NCM, 44 patients had symptomatic neurological complications. Eleven patients developed hydrocephalus, 10 had CNS melanoma, and required ventriculoperitoneal shunt placement. Nine of the 10 patients ultimately died of their disease. Three patients were diagnosed with intraperitoneal melanoma, though data are unavailable for the remaining eight. CONCLUSIONS: All (n=11) patients with NCM-associated CNS melanoma required VP shunts for symptomatic relief. Ten of these patients died within 4.3 years of VP shunt placement, with a range of 1 month to 13.5 years prior to succumbing to their disease. While the intraperitoneal pathology remains unknown for 7 of the cases, 3 had confirmed intraperitoneal melanoma, suggesting that VP shunts provided the conduit to CNS melanoma seeding of the peritoneum. Obtaining baseline abdominal imaging studies prior to VP shunt placement may be helpful in the follow-up of these patients.
- Published
- 2022
25. Phase 2 of trametinib plus radioiodine in RAS-mutant and wild-type, radioiodine-refractory thyroid cancer (ETCTN9446)
- Author
-
Bharat Burman, R. Michael Tuttle, Ravinder K Grewal, Eric Jeffrey Sherman, Shrujal S. Baxi, Laura Boucai, Mona Sabra, Stephanie Fish, Keith S. Pentlow, Sofia Haque, Irina Ostrovnaya, Ronald A Ghossein, Helen X. Chen, John Humm, Michael Anthony Carducci, Steven M. Larson, David G. Pfister, James A Fagin, and Alan Loh Ho
- Subjects
Cancer Research ,Oncology - Abstract
6089 Background: A pilot study showed MEK inhibition could enhance radioiodine (RAI) avidity/efficacy in 5 RAS mutant (MUT), RAI-refractory (RAIR) thyroid cancer (TC) patients (pts). This phase 2 trial with the MEK 1/2 inhibitor trametinib (tram) was conducted to define the efficacy of this “redifferentiation” strategy in RAS MUT RAIR pts and separately in a RAS wild-type (WT) cohort. Methods: Recurrent and/or metastatic, RAIR TC pts w/ RAS MUT (Cohort A) or RAS WT (excluding BRAFV600E) (Cohort B) tumors were treated w/ tram (2 mg orally daily). Progressive disease or new/worsening disease-related symptoms was required for Cohort A pts. 124I PET was performed at baseline and the fourth week of tram. If the second 124I PET showed increased RAI avidity allowing > 2000 cGy to be delivered to a tumor w/ < 300 mCi 131I, pts were treated w/ 131I, guided by whole body and blood dosimetry. Tram was continued through 2 days s/p 131I. Pts who did not qualify for 131I from A/B were taken off study or continued tram alone (Cohort C). For Cohort A (n = 25), the two co-primary endpoints were objective response rate (ORR) and progression-free survival (PFS) 6 months (mos) s/p 131I. Observing either >4 pts w/ confirmed complete or partial response (cCR or cPR) or > 9 progression-free at 6 mos would be considered promising. Secondary endpoints were the proportion of pts w/ increased 124I, safety/tolerability of tram and thyroglobulin changes s/p RAI. The Cohort B primary endpoint was the proportion of pts whose tumors exceeded the lesional dosimetry threshold for 131I w/ tram. An exploratory endpoint for Cohort C was best objective response (BOR) w/ tram. Results: 25 RAS MUT pts enrolled in Cohort A. 23 had at least one (> 1) 124I (-) lesion, 21 had >1 124I (+) lesions and 4 pts had tumors lacking any 124I uptake. After tram treatment, 22/25 had increased 124I uptake; 17/23 had 124I (-) tumors convert positive. Importantly, 15/25 (60%) pts had increased 124I uptake and met lesional dosimetry criteria for 131I on tram. Of 14 pts treated w/ 131I, 8 (57%) achieved cPR, 3 (21%) stable disease (SD) and 3 (21%) progression of disease (PD) 6 mos s/p RAI, translating to 32% ORR and 44% 6-month PFS among all 25 pts. Cohort B had 9 pts (4 Class II BRAF alterations, 4 RET rearrangements, 1 STK11 mutation). 3/4 pts w/ Class II BRAF altered tumors qualified for 131I, leading to 1 cPR, 2 SD 6 mos s/p 131I. 1/4 pts w/ RET rearranged tumors qualified for 131I, producing SD at 6 mos. The STK11 MUT pt did not have increased 124I uptake w/ tram. 7 131I-ineligible pts enrolled to continue tram (Cohort C). Two serious adverse events (grade 3 anemia [Cohort A], grade 3 ejection fraction decrease [Cohort C]) and 3 grade 1 blurred vision/decreased visual acuity AEs were related to tram. Conclusions: Trametinib enhanced RAI uptake/efficacy in a subset of RAS MUT and Class II BRAF altered tumors. Further study to define the efficacy and optimal application of this therapeutic strategy is warranted. Clinical trial information: NCT02152995.
- Published
- 2022
26. EPCT-10. DEBIO1347, AN ORAL FGFR INHIBITOR: RESULTS FROM A SINGLE CENTER STUDY IN RECURRENT/REFRACTORY FGFR ALTERED PEDIATRIC GLIOMAS
- Author
-
Katarzyna Ibanez, Matthias A. Karajannis, Ira J. Dunkel, Sofia Haque, Marc Dinkin, Sameer Farouk Sait, Marc K. Rosenblum, Stephen Gilheeney, Stephanie Vitolano, and Tejus Bale
- Subjects
Pilomyxoid astrocytoma ,Cancer Research ,business.industry ,medicine.disease ,Single Center ,Translational/Early Phase Clinical Trials ,Growth velocity ,Oncology ,Refractory ,Fibroblast growth factor receptor ,Glioma ,Partial response ,Cancer research ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
Background Oncogenic driver alterations in FGFR are present in a subset of pediatric gliomas. Debio1347 is an orally available, highly selective FGFR 1–3 inhibitor that had a favorable safety profile and encouraging preliminary clinical activity in an adult phase 1 study. Methods Five children with progressive/refractory CNS tumors harboring an FGFR gene alteration following prior therapy were treated with Debio1347 at Memorial Sloan Kettering Cancer Center on single patient use protocols. Patients were treated using the 20 mg tablet formulation at the adult recommended phase 2 dose (80 mg/1.73 m2 * BSA once daily). Toxicities were graded using CTCAEv5.0 and imaging response assessments were performed every 8–12 weeks. RESULTS All AEs were grade 1–2. Most common treatment-related adverse events were hyperphosphatemia, ALT increased and hypoalbuminemia (4 patients). Two patients met criteria for partial response and two patients had stable disease. A 13 month-old patient with a spinal cord high-grade glioma harboring two FGFR1 mutations (V592M, K687E) had tumor reduction of 91.7% maintained for 12 months. A 26-month-old patient with a pilomyxoid astrocytoma harboring an FGFR1-TACC1 fusion had a tumor reduction of 74.5% maintained for 9 months. Molecular characterization of recurrent tumor from this patient demonstrated an NF1 deletion as a novel molecular mechanism of acquired resistance to FGFR inhibition. Prolonged disease stabilization was noted in an eight year-old patient with metastatic suprasellar pilomyxoid astrocytoma harboring an FGFR1 mutation (9 months) and in a 14 year-old patient with posterior fossa glioneuronal tumor harboring an FGFR3-TACC3 fusion (24 months and ongoing). Conclusions Debio1347 demonstrated tolerable toxicity and promising anti-tumor efficacy in pediatric patients with refractory FGFR altered gliomas. Specific attention to growth velocity and clinical symptoms with incorporation of imaging assessment of bone growth is warranted. Candidate biomarkers (FGFR1 V592M and K687E SNVs, FGFR-TACC fusions) may guide patient selection. Further studies in this population are warranted.
- Published
- 2021
27. CTNI-33. DEBIO1347, AN ORAL FGFR INHIBITOR: RESULTS FROM A SINGLE CENTER STUDY IN PEDIATRIC PATIENTS WITH RECURRENT/REFRACTORY FGFR ALTERED GLIOMAS
- Author
-
Katarzyna Ibanez, Sofia Haque, Marc K. Rosenblum, Daniel E. Prince, Matthias A. Karajannis, Tejus Bale, Stephen Gilheeney, Ira J. Dunkel, Krisoula Spatz, and Sameer Farouk Sait
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Clinical Trials: Non-Immunologic ,Single Center ,medicine.disease ,Hyperphosphatemia ,Refractory ,Fibroblast growth factor receptor ,Internal medicine ,Glioma ,Troponin I ,medicine ,Neurology (clinical) ,Hypoalbuminemia ,Adverse effect ,business - Abstract
BACKGROUND Oncogenic driver alterations in fibroblast growth factor receptors (FGFRs) are present in a subset of pediatric gliomas. Debio1347 is an orally available, highly selective FGFR 1–3 inhibitor with a favorable safety profile and encouraging preliminary clinical activity in an adult phase 1 study. METHODS We treated five children with progressive/refractory CNS tumors harboring an FGFR gene alteration with Debio1347 on single patient use protocols. Patients were treated using the 20 mg tablet formulation at the adult recommended phase 2 dose (80 mg/1.73 m2 ′ BSA once daily). RESULTS All adverse events (AEs) were grade 1–2. Most common treatment-related AEs were hyperphosphatemia, ALT elevation and hypoalbuminemia. Two patients met criteria for partial response and two patients had stable disease. A 13-month-old patient with a spinal cord high-grade glioma harboring two FGFR1 mutations had tumor reduction of 96.3% maintained for 11 months. A 26 month-old patient with a pilomyxoid astrocytoma harboring an FGFR1-TACC1 fusion had a tumor reduction of 74.5% maintained for 9 months. Prolonged disease stabilization and clinically significant improvement in visual function was noted in an eight year-old patient with metastatic suprasellar pilomyxoid astrocytoma harboring two FGFR1 mutations (14 months overall and sustained for 6 months off therapy) and in a 14 year-old patient with posterior fossa glioneuronal tumor harboring an FGFR3-TACC3 fusion (26 months and ongoing). CONCLUSIONS FGFR targeted therapy with Debio1347 demonstrated tolerable toxicity and promising anti-tumor efficacy in pediatric patients with recurrent/refractory FGFR altered gliomas. Further studies in this population are warranted.
- Published
- 2020
28. Debio1347, an Oral FGFR Inhibitor: Results From a Single-Center Study in Pediatric Patients With Recurrent or Refractory FGFR-Altered Gliomas
- Author
-
Stephen Gilheeney, Marc Dinkin, Krisoula Spatz, Matthias A. Karajannis, Sameer Farouk Sait, Ira J. Dunkel, Katarzyna Ibanez, Sofia Haque, Tejus Bale, Daniel E. Prince, Stephanie Vitolano, and Marc K. Rosenblum
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Adolescent ,business.industry ,Brain Neoplasms ,Infant ,Glioma ,Case Reports ,Single Center ,Receptors, Fibroblast Growth Factor ,Text mining ,Refractory ,Fibroblast growth factor receptor ,Internal medicine ,Child, Preschool ,medicine ,Humans ,Pyrazoles ,Benzimidazoles ,business ,Child - Published
- 2020
29. A Secondary Mutation in BRAF Confers Resistance to RAF Inhibition in a BRAFV600E-Mutant Brain Tumor
- Author
-
Christine A. Pratilas, Sharmeen Uddin, Amy Allen, Zhan Yao, Jiawan Wang, Ira J. Dunkel, Barry S. Taylor, Neal Rosen, Philip Jonsson, Alice Can Ran Qin, Katia Manova, Sofia Haque, David J. Pisapia, Mary Petriccione, and Marc K. Rosenblum
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Mutation ,biology ,business.industry ,medicine.medical_treatment ,Mutant ,Dabrafenib ,Drug resistance ,medicine.disease_cause ,Receptor tyrosine kinase ,Targeted therapy ,03 medical and health sciences ,030104 developmental biology ,Oncology ,medicine ,Cancer research ,biology.protein ,business ,V600E ,medicine.drug - Abstract
BRAFV600E hyperactivates ERK and signals as a RAF inhibitor–sensitive monomer. Although RAF inhibitors can produce impressive clinical responses in patients with mutant BRAF tumors, the mechanisms of resistance to these drugs are incompletely characterized. Here, we report a complete response followed by clinical progression in a patient with a BRAFV600E-mutant brain tumor treated with dabrafenib. Whole-exome sequencing revealed a secondary BRAFL514V mutation at progression that was not present in the pretreatment tumor. Expressing BRAFV600E/L514V induces ERK signaling, promotes RAF dimer formation, and is sufficient to confer resistance to dabrafenib. Newer RAF dimer inhibitors and an ERK inhibitor are effective against BRAFL514V-mediated resistance. Collectively, our results validate a novel biochemical mechanism of RAF inhibitor resistance mediated by a secondary mutation, emphasizing that, like driver mutations in cancer, the spectrum of mutations that drive resistance to targeted therapy are heterogeneous and perhaps emerge with a lineage-specific prevalence. Significance: In contrast to receptor tyrosine kinases, in which secondary mutations are often responsible for acquired resistance, second-site mutations in BRAF have not been validated in clinically acquired resistance to RAF inhibitors. We demonstrate a secondary mutation in BRAF (V600E/L514V) following progression on dabrafenib and confirm functionally that this mutation is responsible for resistance. Cancer Discov; 8(9); 1130–41. ©2018 AACR. See related commentary by Romano and Kwong, p. 1064. This article is highlighted in the In This Issue feature, p. 1047
- Published
- 2018
30. Central nervous system neuroblastoma metastases pseudoprogression following intraventricular anti-B7-H3 radioimmunotherapy
- Author
-
Kim Kramer, Sofia Haque, and Osman Khan
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Neurology ,business.industry ,medicine.medical_treatment ,Central nervous system ,medicine.disease ,Article ,medicine.anatomical_structure ,Internal medicine ,Radioimmunotherapy ,Neuroblastoma ,medicine ,Neurology (clinical) ,business ,Pseudoprogression - Published
- 2019
31. EPCT-21. NEXT-GENERATION SEQUENCING OF CEREBROSPINAL FLUID FOR CLINICAL MOLECULAR DIAGNOSTICS IN ADOLESCENT AND YOUNG ADULT (AYA) BRAIN TUMOR PATIENTS
- Author
-
Onur Yildirim, Irene Rodriguez-Sanchez, Arti Patel, Johnathan Rafailov, Kim Kramer, Alexandra Miller, Nancy Bouvier, Michael F. Berger, Ira J. Dunkel, Sameer Farouk Sait, Matthias A. Karajannis, Jeffrey P. Greenfield, Katherine Hill, Yasmin Khakoo, Hamza Ahmed, Alex Lee, Tejus Bale, Ingo K. Mellinghoff, Luca Szalontay, Ryma Benayed, Mark M. Souweidane, Maria Donzelli, Maria E. Arcila, Sofia Haque, Stephen Gilheeney, and Audrey Mauguen
- Subjects
Medulloblastoma ,Oncology ,Ependymoma ,Cancer Research ,medicine.medical_specialty ,business.industry ,Brain tumor ,medicine.disease ,Molecular diagnostics ,Translational/Early Phase Clinical Trials ,Cerebrospinal fluid ,Cell-free fetal DNA ,Internal medicine ,Glioma ,medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Young adult ,business - Abstract
Purpose Pediatric central nervous system tumors remain a leading cause of cancer-related death in children and adolescents. Safe sampling of tumor tissue for diagnostic purposes may be challenging. Subclinical detection of disease prior to clinical or imaging progression may provide opportunity for earlier intervention and ultimately improve overall survival. Additionally, our understanding of molecular evolution in response to therapy remains limited, given the rarity of serial sampling of tumor tissue. Methods We report our experience with minimally invasive molecular diagnostics using a validated next generation sequencing assay for sequencing of cerebrospinal fluid (CSF) cell-free DNA (cfDNA) obtained at the time of surgery, by intraventricular catheter or lumbar puncture. All CSF samples were collected as part of clinical care, and results reported to both clinicians and patients/families. Results We analyzed 64 CSF samples from 45 pediatric and adolescent and young adult (AYA) patients (pediatric=25; AYA=20) with primary and recurrent brain tumors across 12 histopathological subtypes including high-grade glioma (n=10), medulloblastoma (n=10), pineoblastoma (n=5), low grade glioma (n=4), diffuse leptomeningeal glioneuronal tumor (DLGNT) (n=4), metastatic retinoblastoma (n=4), ependymoma (n=3), and other (n=5). Somatic alterations were detected in 28/64 samples (44.4%) and in at least one sample per unique patient in 22/45 patients (48.8%). CSF cfDNA positivity was strongly associated with the presence of disseminated disease at the time of collection (86.3%). No association was seen between CSF cfDNA positivity and the timing of CSF collection during the patient’s disease course. Conclusion We identified four general categories where CSF cfDNA testing provided additional relevant diagnostic, prognostic, and/or therapeutic information, impacting clinical assessment and decision making: 1) diagnosis; 2) identification of actionable alterations; 3) track response to therapy; and 4) monitoring tumor evolution. Our findings support broader implementation of clinical CSF cfDNA testing in this population that may improve care.
- Published
- 2021
32. A phase II study of temsirolimus added to low-dose weekly carboplatin and paclitaxel for patients with recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC)
- Author
-
Shrujal S. Baxi, Lara Dunn, Nora Katabi, Eric J. Sherman, C. Pfister, Alan Loh Ho, Matthew G. Fury, Sofia Haque, Han Xiao, Susan H. Korte, and David G. Pfister
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Paclitaxel ,medicine.medical_treatment ,Phases of clinical research ,Drug Administration Schedule ,Carboplatin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Progression-free survival ,Neoplasm Metastasis ,Aged ,Aged, 80 and over ,Sirolimus ,Chemotherapy ,Dose-Response Relationship, Drug ,Squamous Cell Carcinoma of Head and Neck ,business.industry ,Original Articles ,Hematology ,Middle Aged ,medicine.disease ,Corrigenda ,Chemotherapy regimen ,Head and neck squamous-cell carcinoma ,Temsirolimus ,030104 developmental biology ,chemistry ,Tolerability ,Head and Neck Neoplasms ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Female ,Neoplasm Recurrence, Local ,business ,medicine.drug - Abstract
Background Activating events along the PI3K/mTOR pathway are common in head and neck squamous cell carcinomas (HNSCC), and preclinical studies suggest additive or synergistic effects when combining mTORC1 inhibitors with carboplatin and paclitaxel chemotherapy. Patients and methods In this single-institution phase II study, the combination of temsirolimus 25 mg, carboplatin AUC 1.5, and paclitaxel 80 mg/m2 administered on days 1 and 8 of a 21-day cycle was evaluated in 36 patients with recurrent and/or metastatic (R/M) HNSCC. The primary end point was objective response rate after two cycles of treatment. Secondary end points include the safety and tolerability profile and overall survival. Correlative studies with exome mutational analysis were performed in pre-treatment biopsy samples from 21 patients. Results Fifteen (41.7%) patients had an objective response, which were all partial responses, and 19 (52.3%) patients had stable disease as best response. The two patients who were designated as ‘non-responders’ were removed from study prior to two cycles of treatment, but are included in the efficacy and safety analyses. The median duration on study was 5.3 months and the median progression-free survival and overall survival were 5.9 months (95% confidence interval, 4.8–7.1) and 12.8 months (95% confidence interval, 9.8–15.8), respectively. The most common grade 3 and 4 adverse events were hematologic toxicities. Three (3.8%) patients developed neutropenic fever on study. Three of four patients with PIK3CA mutations experienced tumor regressions, and responses were also seen in patients with other genetic alterations in the PI3K/mTOR pathway. Conclusion The combination of temsirolimus with low-dose weekly carboplatin and paclitaxel appears to have meaningful clinical efficacy in the treatment of R/M HNSCC. This regimen has a relatively high response rate compared to other treatments evaluated in R/M HNSCC, and potential associations with genetic alterations in the PI3K/mTOR pathway should be further explored.
- Published
- 2017
33. Phase 2 study evaluating the combination of sorafenib and temsirolimus in the treatment of radioactive iodine-refractory thyroid cancer
- Author
-
Ronald Ghossein, Grace Cullen, Alan L. Ho, James A. Fagin, Eric J. Sherman, Matthew G. Fury, Shrujal S. Baxi, Lara Dunn, Sofia Haque, Cami S. Sima, and David G. Pfister
- Subjects
0301 basic medicine ,Sorafenib ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Temsirolimus ,Thyroid carcinoma ,03 medical and health sciences ,Regimen ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Mucositis ,Anaplastic thyroid cancer ,business ,Survival rate ,Thyroid cancer ,medicine.drug - Abstract
Background Patients with recurrent and/or metastatic, radioactive iodine-refractory thyroid carcinoma have limited treatment options. Sorafenib, an oral kinase inhibitor, is approved by the US Food and Drug Administration for the treatment of radioactive iodine-refractory thyroid carcinoma, although it demonstrated low response rates (12.2%) as a single agent in the first-line setting. The objective of the current study was to determine whether adding the mammalian target of rapamycin inhibitor temsirolimus to sorafenib could improve on these results. Methods In this single-institution, phase 2 study, 36 patients with metastatic, radioactive iodine-refractory thyroid carcinoma of follicular origin received treatment with the combination of oral sorafenib (200 mg twice daily) and intravenous temsirolimus (25 mg weekly). The receipt of prior systemic treatment with cytotoxic chemotherapy and targeted therapy, including sorafenib, was permitted. The primary endpoint was the radiographic response rate. Results The best response was a partial response in 8 patients (22%), stable disease in 21 (58%), and progressive disease in 1 (3%). Six patients were not evaluable for a response. Patients who had received any prior systemic treatment had a response rate of 10% compared with 38% of those who had not received prior systemic treatment. One of 2 patients with anaplastic thyroid cancer had an objective response. The progression-free survival rate at 1 year was 30.5%. The most common grade 3 and 4 toxicities associated with sorafenib and temsirolimus included hyperglycemia, fatigue, anemia, and oral mucositis. Conclusions Sorafenib and temsirolimus appear to be an active combination in patients with radioactive iodine-refractory thyroid carcinoma, especially in patients who received no prior treatment compared with historic data from single-agent sorafenib. Activity is also observed in patients who previously received sorafenib. This regimen warrants further investigation. Cancer 2017;123:4114-4121. © 2017 American Cancer Society.
- Published
- 2017
34. NCOG-60. MALIGNANT MELANOMA IN NEUROCUTANEOUS MELANOCYTOSIS: A RETROSPECTIVE CASE SERIES (2000-2020)
- Author
-
Ugur Sener, Elsie Ennin, Stephanie Suser, Yasmin Khakoo, Sofia Haque, Ashfaq A. Marghoob, and Kevin Elmore
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,Melanoma ,medicine ,Neurology (clinical) ,medicine.disease ,business ,Dermatology ,Neurocutaneous melanocytosis ,Outcome Measures and Neuro-Cognitive Outcomes - Abstract
Neurocutaneous melanocytosis (NCM) is a rare neurocutaneous syndrome which typically develops in children with large congenital melanocytic nevi (LCMN) and excessive melanocyte proliferation in the leptomeninges and brain parenchyma. Malignant melanoma develops in an estimated 2.3% of patients with LCMN and 40-60% of patients with NCM. NCM-associated melanomas frequently harbor NRAS mutations with no well-established role for targeted therapy. In a retrospective, single-institution study, we reviewed eleven patients with NCM-associated CNS melanoma evaluated at Memorial Sloan Kettering Cancer Center from June 2000 to January 2020. Five patients had previously identified focal melanocytosis prior to developing melanoma. In this subgroup, the median time from identification of focal melanocytosis to melanoma diagnosis was 80 months (range: 18-200). Median age at melanoma diagnosis was 9.9 years (range: 1.1-25.3). Presentation at the time of diagnosis with CNS melanoma included headache (45%), focal deficits (45%), and seizure (18%). Eight patients had hydrocephalus (73%). Five patients presented with a focal mass (45%) and six patients had focal or diffuse leptomeningeal disease without a mass (55%). Leptomeningeal spread eventually developed in all patients. Where molecular testing was available, three melanomas had NRAS mutations and none were associated with BRAF mutations. Seven patients were treated with cancer-directed therapy including temozolomide, trametinib, ipilimumab, and nivolumab, with each therapy being administered to two patients. Radiation therapy was used in three patients, including whole brain radiation therapy and stereotactic radiosurgery. Median survival from melanoma diagnosis was 9.1 months (range: 1-60.4). The longest surviving patient was initially diagnosed with cutaneous melanoma, surviving 60.4 months after diagnosis with cutaneous melanoma and 22.7 months after diagnosis with CNS melanoma. Prognosis remains guarded in patients with NCM-associated melanoma, and further investigation is warranted to identify effective management strategies.
- Published
- 2020
35. Reliability of CT myelography versus MRI in the assessment of spinal epidural disease
- Author
-
Julio Arevalo-Perez, Andrei I. Holodny, Kyung K. Peck, Lillian C. Chen, Sasan Karimi, Yoshiya Yamada, John K. Lyo, Jamie Tisnado, Eric Lis, and Sofia Haque
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,Soft tissue ,Magnetic resonance imaging ,medicine.disease ,Sagittal plane ,Article ,Radiation therapy ,Degenerative disease ,medicine.anatomical_structure ,Cerebrospinal fluid ,medicine ,Radiology, Nuclear Medicine and imaging ,Radiology ,Radiation treatment planning ,business ,Myelography - Abstract
The spine is the third most common site of metastatic disease after the lungs and liver [1–2]. Several imaging modalities exist for the evaluation of epidural disease, namely computed tomography (CT) and magnetic resonance imaging (MRI). CT myelography (CTM) involves the injection of contrast material into the cerebrospinal fluid prior to imaging and requires ionizing radiation. Though previously widely used, CTM has steadily fallen out of favor to MRI at many institutions and in some cases, is used alone in radiation therapy planning. MRI is notable for its non-invasive nature, excellent visualization of soft tissues, and lack of exposure to ionizing radiation. CTM, however, can provide improved visualization of bony structures and is more resistant than MRI to geometric distortion and patient motion due to the rapid acquisition of images [3]. Additionally, CTM can be used in patients with contraindications to MRI and can also provide a direct correlation of electron density to doses used for radiation therapy planning [4]. With recent advances in radiation therapy, including intensity-modulated and image-guided radiation therapy, which aim to improve tumor targeting thereby sparing normal tissue, precise tumor localization for treatment planning often requires the use of CT integrated with MRI [5–7]. A previous study comparing CTM and MRI in the evaluation of degenerative disease in the cervical spine found that MRI was superior in its reliability for assessing the degree of nerve root compression while CTM provided greater reliability in the assessment of bony lesions and foraminal stenosis [8]. Other studies focusing on the lumbar spine also found that CTM provided greater reliability and reproducibility in the assessment of the extent of lumbar stenosis than did MRI [9] and that MRI underestimated the degree of spinal root compression in the lumbar spine [10]. While both imaging modalities have complementary advantages, studies to date have not investigated a composite evaluation of spinal epidural disease. Furthermore, given the limitations of MRI, it has not been studied whether there is merit in continuing to use CTM to complement MRI for radiation therapy planning. This study is the first to investigate whether axial and sagittal projections of the spinal epidural space using both T1 and T2-weighted MRI compared to CTM produce compatible levels of inter- and intra-rater reliability in measuring the extent of epidural disease by neuroradiologists trained in both imaging modalities. The results of this study will be useful in determining whether CTM should continue to be used in conjunction with MRI when evaluating spinal metastases and planning radiation therapy.
- Published
- 2019
36. Sustained imaging response and hearing preservation with low-dose bevacizumab in sporadic vestibular schwannoma
- Author
-
Sofia Haque, Mari Hagiwara, Matthias A. Karajannis, and Mark Schreyer
- Subjects
Cancer Research ,medicine.medical_specialty ,Bevacizumab ,Hearing loss ,Schwannoma ,Antineoplastic Agents, Immunological ,Medicine ,Humans ,Letters to the Editor ,Hearing Loss ,Vestibular system ,Hearing preservation ,business.industry ,Low dose ,Neuroma, Acoustic ,Middle Aged ,medicine.disease ,Neuroma ,Prognosis ,Magnetic Resonance Imaging ,Oncology ,Female ,Neurology (clinical) ,Radiology ,medicine.symptom ,business ,medicine.drug - Published
- 2019
37. LGG-02. A PHASE II PROSPECTIVE TRIAL OF SELUMETINIB IN CHILDREN WITH RECURRENT/PROGRESSIVE PEDIATRIC LOW-GRADE GLIOMA (PLGG) WITH A FOCUS UPON OPTIC PATHWAY/HYPOTHALAMIC TUMORS AND VISUAL ACUITY OUTCOMES: A PEDIATRIC BRAIN TUMOR CONSORTIUM (PBTC) STUDY, PBTC-029B
- Author
-
Azra H. Ligon, Lindsay Kilburn, Tina Young Poussaint, Ibrahim Qaddoumi, Maryam Fouladi, Michael Fisher, Regina I. Jakacki, Gilbert Vezina, Malcolm G. Smith, Neal I. Lindeman, Clinton F. Stewart, Zoltan Patay, Ashok Panigrahy, David T.W. Jones, Ira J. Dunkel, Patricia Baxter, Stewart Goldman, Girish Dhall, Susa G Kreissman, Sofia Haque, Austin Doyle, Roger J. Packer, David S. Enterline, Jason Fangusaro, Arzu Onar-Thomas, Paul G. Fisher, Jeremy Jones, Blaise V. Jones, Soonme Cha, Benita Tamrazi, Anu Banerjee, Shengjie Wu, Ian F. Pollack, Stefan M. Pfister, and Jessica S Stern
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Visual acuity ,Pediatric Brain Tumor Consortium ,business.industry ,Phases of clinical research ,Low Grade Glioma ,medicine.disease ,Internal medicine ,Glioma ,medicine ,Selumetinib ,Hypothalamic Neoplasm ,Low-Grade Glioma ,Neurology (clinical) ,Progression-free survival ,medicine.symptom ,business - Abstract
BACKGROUND: Pediatric low-grade glioma (pLGG) is the most common CNS tumor of childhood. Progression-free survival (PFS) is much lower than overall survival emphasizing the need for alternative treatments. In addition, many children suffer functional morbidities such as visual and motor disturbances. Recently, there has been an appropriate prioritization of functional outcomes in children with pLGG. METHODS: We present the results of a PBTC phase II trial evaluating selumetinib, (AZD6244, ARRY-142886) a MEK-1/2 inhibitor, in children with recurrent/progressive pLGG on 2 strata, including visual outcomes. RESULTS: Stratum 3 enrolled Neurofibromatosis type 1 (NF1)-associated pLGG. Ten of 25 (40%) eligible patients had partial response (PR), 14/25 (56%) had stable disease (SD) and 1/25 (4%) had progressive disease (PD); 2-year PFS was 96+4%. Ten patients with optic pathway glioma (OPG) were evaluable for visual acuity (VA) at baseline and 1 year. VA improved in 2/10 patients (20%) and was stable in 8/10 (80%). One patient (10%) had improvement in visual fields (VF) and 9 patients (90%) had stable VF. Stratum 4 included patients with non-NF1-associated recurrent/progressive hypothalamic and OPG. Five of 25 (20%) eligible patients had PR, 16/25 (64%) had SD and 4 (16%) had PD; 2-year PFS was 78+8.5%. Nineteen of 25 patients were evaluable for VA. VA improved in 4/19 patients (21%), was stable in 13/19 (68%) and worsened in 2/19 (11%). Five patients (26%) had improved VF and 14 (74%) had stable VF. The most common toxicities included grade 1/2 CPK elevation, diarrhea, hypoalbuminemia and rash. Rare grade 3 toxicities included elevated CPK, rash and paronychia. CONCLUSIONS: Selumetinib was tolerable and effective in treating children with NF1-associated and sporadic recurrent/progressive hypothalamic and OPG based upon radiographic response and PFS. Twenty-seven of 29 (93%) evaluable patients had stable or improved vision based on VA and VF testing.
- Published
- 2019
38. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial
- Author
-
Girish Dhall, Laurence Austin Doyle, Lindsay Kilburn, Soonmee Cha, Sofia Haque, Susan G. Kreissman, David S. Enterline, Ira J. Dunkel, Roger J. Packer, Jason Fangusaro, Clinton F. Stewart, Zoltan Patay, Maryam Fouladi, David T.W. Jones, Shengjie Wu, Ashok Panigrahy, Michael Fisher, Ibrahim Qaddoumi, Patricia Baxter, Regina I. Jakacki, Benita Tamrazi, Neal I. Lindeman, Azra H. Ligon, Anuradha Banerjee, Stewart Goldman, Tina Young Poussaint, Arzu Onar-Thomas, Malcolm A. Smith, Ian F. Pollack, Stefan M. Pfister, Gilbert Vezina, Paul G. Fisher, Jeremy Jones, and Jessica S Stern
- Subjects
Male ,Phases of clinical research ,Central Nervous System Neoplasms ,Neoplasms, Multiple Primary ,0302 clinical medicine ,Multiple Primary ,Neoplasms ,Clinical endpoint ,Maculopapular rash ,Child ,Cancer ,Pediatric ,Pilocytic astrocytoma ,Glioma ,Oncology ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,Child, Preschool ,Disease Progression ,Female ,medicine.symptom ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,Neurofibromatosis 1 ,Adolescent ,Pediatric Cancer ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Article ,03 medical and health sciences ,Young Adult ,Rare Diseases ,Clinical Research ,Internal medicine ,medicine ,Humans ,Oncology & Carcinogenesis ,Adverse effect ,Preschool ,business.industry ,Neurosciences ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Brain Disorders ,Clinical trial ,Brain Cancer ,Selumetinib ,Benzimidazoles ,Neoplasm Grading ,business ,030217 neurology & neurosurgery - Abstract
BACKGROUND: Pediatric low-grade glioma (pLGG) is the most common central nervous system tumor of childhood. Although overall survival is very good, many children suffer from multiple progressions and functional morbidities. There is no one universally accepted therapy for children with recurrent disease, however, standard cytotoxic chemotherapies are often utilized by most practitioners. The Pediatric Brain Tumor Consortium conducted a multi-institutional phase II study evaluating selumetinib (AZD6244, ARRY-142886), a MAP/ERK Kinase I/II inhibitor, in patients with recurrent, refractory or progressive pLGG assigned to numerous strata. The aim of the study was to evaluate the efficacy of selumetinib in these patients. METHODS: Eligibility required age 3–21 y/o, a Lansky or Karnofsky performance score greater than 60 and the presence of recurrent, refractory or progressive pLGG after at least one standard therapy. Stratum 1 included children with World Health Organization (WHO) grade I pilocytic astrocytoma (PA) harboring either one of the two most common BRAF aberrations (KIAA1549-BRAF fusion or the BRAF(V600E) mutation). Stratum 3 included children with any neurofibromatosis type 1 (NF1)-associated pLGG (WHO grades I and II). Selumetinib was provided as capsules given orally at the recommended phase II dose of 25 mg/m(2) twice daily. The primary endpoint was stratum-specific objective response rate assessd by the local site and sustained for at least 8 weeks. All responses were reviewed centrally and statistical analyses were done as per protocol. Although the trial (NCT01089101) is still ongoing in other strata, enrollment and planned follow-up is compete on both strata 1 and 3. FINDINGS: Between July 25, 2013, and June 12, 2015, 25 eligible and evaluable children were accrued to stratum 1, and between August 28, 2013, and June 25, 2015, 25 eligible and evaluable children were accrued to stratum 3. On stratum 1, 9/25 (36%) patients achieved a partial response (PR). The median follow-up for the 11 patients who have not yet experienced an event is 36.4 months (4.4–50.5; IQR=23.9). On stratum 3, 10/25 (40%) patients achieved a PR with a median follow-up of 48.6 months (8.6–59.1; IQR=12.2) for the 17 subjects without progressions. All patients evaluable for visual acuity had improved or stable vision. The most common attributable toxicities on both strata were grade 1 and 2 CPK elevation, hypoalbuminemia, dyspnea, rash, duodenal ulcer, anemia, dry skin, fatigue and diarrhea. Rare grade 3 toxicities included elevated CPK (n=5), maculopapular rash (n=5), neutropenia (n=3), nausea (n=3), paronychia (n=3), acneiform rash (n=2), diarrhea (n=2), elevated ALT (n=1), decreased ejection fraction (n=1), gastric hemorrhage (n=1), headache (n=1), skin infection (n=1), tooth infection (n=1) and weight gain (n=1). There was only one grade 4 toxicity, lymphopenia. There were no treatment-realted deaths. Patient reported outcomes and quality of life assessments were not part of the current study. INTERPRETATION: Selumetinib is active against recurrent, refractory or progressive PA harboring common BRAF aberrations and NF1-associated pLGG. To our knowledge, this is one of the first prospectively tested and successful molecularly-targeted agents in pLGG. These data not only provide an alternative to standard chemotherapy for these subgroups of patients, but this success has led to an interest in exploring efficacy in patients as a first-line therapy. In fact, these data have directly led to the development of two Children’s Oncology Group phase III studies in newly diagnosed pLGG patients both with and without NF1 comparing standard chemotherapy to selumetinib. The current trial was funded by a National Cancer Institute (NCI) Cancer Therapy Evaluation Program (CTEP) PBTC U01 Grant: 2UM1CA081457 (UM1) and by the American Lebanese Syrian Associated Charities.
- Published
- 2019
39. Phase 1 dose-escalation trial using convection-enhanced delivery of radiolabeled monoclonal antibody for diffuse intrinsic pontine glioma following external radiation therapy
- Author
-
John Roemer Nielsen, Ira J. Dunkel, Serge K. Lyashchenko, Mark M. Souweidane, Maria Donzelli, Jorge A. Carrasquillo, Neeta Pandit-Taskar, Kim Kramer, Yasmin Khakoo, Pat Zanzonico, Steven M. Larson, Sofia Haque, Sunitha B. Thakur, Jason S. Lewis, and Nai-Kong V. Cheung
- Subjects
Cancer Research ,Oncology ,medicine.drug_class ,business.industry ,Cancer research ,medicine ,Dose escalation ,Overall survival ,External Radiation Therapy ,Monoclonal antibody ,business ,Convection-Enhanced Delivery - Abstract
2010 Background: The prognosis of diffuse intrinsic pontine glioma (DIPG) is dire with a median overall survival less than one-year. 124I-omburtamab is a radiolabeled monoclonal antibody that targets B7-H3 epitope. We evaluated the safety of administering escalating doses and volumes of 124I-omburtamab via convection-enhanced delivery (CED) in children with DIPG. Methods: MSKCC 11-011 trial is a standard 3+3 phase 1, open-label, dose escalation study in patients with non-progressive DIPG. CED of 124I-omburtamab was performed between 4-14 weeks post-external radiation therapy. Nine dose levels of a single injection of 124I-omburtamab (Y-mAbs Therapeutics, USA) (range 0.25 to 8.0 mCi; and volume of infusion (Vi) from 250 to 8,000 µl) have been evaluated so far. Patients were assessed weekly for 30 days. Results: 46 children were evaluable for primary and secondary endpoints. The median age at enrolment was 6.5 years (range 2-17). Two patients have experienced AEs CTCAE grade 3 that were categorized as dose limiting toxicities (DLTs), which led to inclusion of three more patients at both the 4 and 6 mCi dose levels. Eight patients have reported transient AEs of grade 3 considered related to 124I-omburtamab. The acute grade 3 AEs were generally indicative of nervous system effects due to volume intolerance or radiation injury, and included hemiparesis (n = 3), dysarthria (n = 3), ataxia (n = 3), dysphagia (n = 2), muscular weakness (n = 2) and gait disturbance (n = 1). There were no related AEs CTCAE grade 4 or 5. Estimations of distribution volumes based on T2-weighted imaging were linearly related to volume with a mean volume of distribution/volume of infusion ratio (Vd/Vi) between 3 and 3.5. The mean ratio of lesion-to-whole body absorbed dose was ̃1000. Median overall survival from diagnosis across all cohorts was 14.8 months (n = 46, 95% CI 11.5, 16.8) and the survival rate estimates (with 95% confidence intervals) at 1, 2, 3 and 5 years were 0.63 (0.46;0.76); 0.13 (0.05;0.26); 0.08 (0.02;0.19); and 0.04 (0.00;0.16), respectively. Four patients have survived > 3 years; two remain alive at 46 and 96 months and two have died at 43 and 53 months, both with CNS disease outside of the treatment field and one with extra-CNS metastases. Conclusions: 124I-omburtamab via CED into the brain stem of children with DIPG and previously irradiated provides a possibility for improved treatment of DIPG. A dose of 8mCi and an infusion volume of 8,000 µl is considered safe and may provide a distribution volume large enough to cover tumor volumes up to 20 cm3. The median overall survival of all patients included in the trial appears to be increased with 3-4 months compared to historical control data from consortia trials. A phase 2 trial aiming at investigating the efficacy of radiolabeled omburtamab administered via CED is being planned. Clinical trial information: NCT01502917.
- Published
- 2021
40. A phase II trial cohort of nivolumab plus ipilimumab in patients (Pts) with recurrent/metastatic salivary gland cancers (R/M SGCs)
- Author
-
Irina Ostrovnaya, Eric J. Sherman, Sofia Haque, Luc G. T. Morris, David G. Pfister, Bharat Burman, Lara Dunn, James Vincent Fetten, Loren S. Michel, and Alan Loh Ho
- Subjects
Minor Salivary Glands ,Cancer Research ,medicine.medical_specialty ,Salivary gland ,business.industry ,Standard treatment ,Ipilimumab ,Gastroenterology ,medicine.anatomical_structure ,Oncology ,Internal medicine ,Cohort ,medicine ,In patient ,Nivolumab ,business ,medicine.drug - Abstract
6002 Background: R/M SGCs are a diverse group of malignant neoplasms arising from the major or minor salivary glands and have no standard treatment. The impact of combining PD-1/CTLA-4 checkpoint blockade in R/M SGCs is unknown. Methods: In a Simon's two-stage minimax phase II trial, pts with progressive R/M SGCs (any histology except adenoid cystic carcinoma (ACC)) were enrolled and treated with nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks (1 cycle = 6 weeks). Imaging, using RECIST v1.1 response assessment, was scheduled to be performed approximately every 12 weeks. The primary endpoint was best overall response (BOR = complete response [CR]+partial response [PR]) per RECIST v1.1. To detect a difference between an unacceptable BOR of 5% and a desirable BOR of 20% (one-sided type I error of 10%, power of 90%), at least 1 in the first 18 pts required an observed response. At least 4 responses of 32 total pts were needed to meet the primary endpoint. Treatment beyond progression of disease (PD) was allowed at the discretion of the investigator. A second cohort of pts with ACC was analyzed and reported separately. Results: From 7/25/2017-7/16/2020, 32 pts were enrolled and evaluable for the primary endpoint. There was 3 confirmed PRs in the first 18 pts, therefore enrollment of the second stage continued. BOR rate was 16% (5/32). Seven pts never reached a first disease assessment and were classified as non-responders: 5 due to clinical PD, 1 due to toxicity, and 1 pt withdrew. Four pts discontinued the trial for toxicities: pancytopenia (1), blurry vision (1), cardiomyopathy/hyperglycemia (1), and neutropenic sepsis (1), and mucositis (1). The 5 confirmed responders had regressions ranging from -66% to -100% in target lesions, with a duration of therapy ranging from 15.7 to 29.5 months (treatment ongoing for one as of 2/6/20). Conclusions: This cohort met its primary endpoint, and the responses observed were dramatic and durable. Paired biopsy and peripheral blood samples will be analyzed to elucidate insights into mechanisms of response and resistance to dual checkpoint blockade. Clinical trial information: NCT03172624.
- Published
- 2021
41. Prelaminar and Postlaminar Invasion of Retinoblastoma
- Author
-
David H. Abramson, Jasmine H. Francis, and Sofia Haque
- Subjects
Male ,medicine.medical_specialty ,Retinoblastoma ,business.industry ,Optic Nerve Neoplasms ,Retinal Neoplasms ,MEDLINE ,Optic Nerve ,medicine.disease ,Ophthalmology ,Child, Preschool ,medicine ,Humans ,Neoplasm Invasiveness ,business ,Ultrasonography - Published
- 2021
42. RARE-24. LARGE CONGENITAL MELANOCYTIC NEVI AND NEUROCUTANEOUS MELANOCYTOSIS: A RETROSPECTIVE CASE SERIES
- Author
-
Elsie Ennin, Ashfaq A. Marghoob, Ugur Sener, Stephanie Suser, Yasmin Khakoo, and Sofia Haque
- Subjects
Cancer Research ,medicine.medical_specialty ,Oncology ,business.industry ,Medicine ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business ,Dermatology ,Neurocutaneous melanocytosis ,Craniopharyngioma and Rare Tumors - Abstract
Neurocutaneous melanocytosis (NCM) is a rare disease characterized by excessive proliferation and deposition of melanocytes in the leptomeninges and brain parenchyma, occurring in children with large congenital melanocytic nevi (LCMN). Manifestations of NCM range from asymptomatic CNS melanin deposition to cranial neuropathies, seizures, and hydrocephalus. Patients with NCM are at risk for malignant melanoma. We conducted a retrospective, single-institution study of patients with LCMN evaluated at Memorial Sloan Kettering Cancer Center from June 2000 to January 2020. Of 55 patients studied, 15 had no radiographic NCM, and 40 had radiographic NCM at initial evaluation. MRI findings included: focal melanocytosis (33), diffuse leptomeningeal disease (4), solid melanoma (3). Malformations were identified in 13, including arachnoid cyst (4), congenital hydrocephalus (4), Dandy-Walker malformation (3), and tethered cord (1). Twenty-one patients completed imaging once and were followed clinically. Seventeen with serial imaging (10 with focal melanocytosis, 7 with normal MRI) remained stable over a median 24-month follow up (range: 1–124). Six had suspected radiographic progression of NCM without melanoma. Malignant melanoma developed in 11 patients, 5 with focal melanocytosis on initial imaging. Median time from focal melanocytosis identification to melanoma diagnosis was 80 months (range: 18–200). Median age at melanoma diagnosis was 9.9 years (range: 1.1–25.3). Median survival from melanoma diagnosis was 9.1 months (range: 1–60.4). Focal NCM on neuroaxis imaging does not predict time to transformation to malignant melanoma. Serial imaging is not indicated in absence of disease-modifying treatment. Clinical follow up of at-risk individuals is essential in early identification of complications.
- Published
- 2020
43. A novel magnetic resonance imaging segmentation technique for determining diffuse intrinsic pontine glioma tumor volume
- Author
-
Robert J. Young, Kyung K. Peck, Sunitha B. Thakur, Zhiping Zhou, Mark M. Souweidane, Sofia Haque, Jamie Tisnado, Ranjodh Singh, and Apostolos John Tsiouris
- Subjects
Male ,Concordance ,Article ,Antibodies, Monoclonal, Murine-Derived ,03 medical and health sciences ,0302 clinical medicine ,Picture archiving and communication system ,medicine ,Animals ,Brain Stem Neoplasms ,Humans ,Segmentation ,Child ,Cluster analysis ,medicine.diagnostic_test ,business.industry ,k-means clustering ,Magnetic resonance imaging ,Glioma ,General Medicine ,Confidence interval ,Tumor Burden ,Diffusion Magnetic Resonance Imaging ,Child, Preschool ,030220 oncology & carcinogenesis ,Polygon ,Female ,business ,Nuclear medicine ,030217 neurology & neurosurgery - Abstract
OBJECTIVE Accurately determining diffuse intrinsic pontine glioma (DIPG) tumor volume is clinically important. The aims of the current study were to 1) measure DIPG volumes using methods that require different degrees of subjective judgment; and 2) evaluate interobserver agreement of measurements made using these methods. METHODS Eight patients from a Phase I clinical trial testing convection-enhanced delivery (CED) of a therapeutic antibody were included in the study. Pre-CED, post–radiation therapy axial T2-weighted images were analyzed using 2 methods requiring high degrees of subjective judgment (picture archiving and communication system [PACS] polygon and Volume Viewer auto-contour methods) and 1 method requiring a low degree of subjective judgment (k-means clustering segmentation) to determine tumor volumes. Lin's concordance correlation coefficients (CCCs) were calculated to assess interobserver agreement. RESULTS The CCCs of measurements made by 2 observers with the PACS polygon and the Volume Viewer auto-contour methods were 0.9465 (lower 1-sided 95% confidence limit 0.8472) and 0.7514 (lower 1-sided 95% confidence limit 0.3143), respectively. Both were considered poor agreement. The CCC of measurements made using k-means clustering segmentation was 0.9938 (lower 1-sided 95% confidence limit 0.9772), which was considered substantial strength of agreement. CONCLUSIONS The poor interobserver agreement of PACS polygon and Volume Viewer auto-contour methods highlighted the difficulty in consistently measuring DIPG tumor volumes using methods requiring high degrees of subjective judgment. k-means clustering segmentation, which requires a low degree of subjective judgment, showed better interobserver agreement and produced tumor volumes with delineated borders.
- Published
- 2016
44. A phase II study of axitinib (AG-013736) in patients with incurable adenoid cystic carcinoma
- Author
-
Cristina R. Antonescu, Camelia Sima, Luc G. T. Morris, David G. Pfister, Ai Ni, Alan Loh Ho, Nora Katabi, Snjezana Dogan, Grace Cullen, Matthew G. Fury, Raghu Chandramohan, Eric J. Sherman, Shrujal S. Baxi, Lara Dunn, and Sofia Haque
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Indazoles ,Axitinib ,Drug-Related Side Effects and Adverse Reactions ,Oncogene Proteins, Fusion ,Adenoid cystic carcinoma ,medicine.drug_class ,Phases of clinical research ,Context (language use) ,Disease-Free Survival ,Tyrosine-kinase inhibitor ,Proto-Oncogene Proteins c-myb ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,MYB ,Protein Kinase Inhibitors ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Imidazoles ,High-Throughput Nucleotide Sequencing ,Original Articles ,Hematology ,Middle Aged ,medicine.disease ,Carcinoma, Adenoid Cystic ,NFI Transcription Factors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Chromosomes, Human, Pair 4 ,business ,Progressive disease ,medicine.drug - Abstract
Background Recurrent/metastatic adenoid cystic carcinoma (ACC) is an incurable disease with no standard treatments. The majority of ACCs express the oncogenic transcription factor MYB (also c-myb), often in the context of a MYB gene rearrangement. This phase II trial of the tyrosine kinase inhibitor (TKI) axitinib (Pfizer) tested the hypothesis that targeting pathways activated by MYB can be therapeutically effective for ACC. Patients and methods This is a minimax two-stage, phase II trial that enrolled patients with incurable ACC of any primary site. Progressive or symptomatic disease was required. Patients were treated with axitinib 5 mg oral twice daily; dose escalation was allowed. The primary end point was best overall response (BOR). An exploratory analysis correlating biomarkers to drug benefit was conducted, including next-generation sequencing (NGS) in 11 patients. Results Thirty-three patients were registered and evaluable for response. Fifteen patients had the axitinib dose increased. Tumor shrinkage was achieved in 22 (66.7%); 3 (9.1%) had confirmed partial responses. Twenty-five (75.8%) patients had stable disease, 10 of whom had disease stability for >6 months. The median progression-free survival (PFS) was 5.7 months (range 0.92–21.8 months). Grade 3 axitinib-related toxicities included hypertension, oral pain and fatigue. A trend toward superior PFS was noted with the MYB/NFIB rearrangement, although this was not statistically significant. NGS revealed three tumors with 4q12 amplification, producing increased copies of axitinib-targeted genes PDGFR/KDR/KIT. Two 4q12 amplified patients achieved stable disease for >6 months, including one with significant tumor reduction and the longest PFS on study (21.8 months). Conclusions Although the primary end point was not met, axitinib exhibited clinical activity with tumor shrinkage achieved in the majority of patients with progressive disease before trial enrollment. Analysis of MYB biomarkers and genomic profiling suggests the hypothesis that 4q12 amplified ACCs are a disease subset that benefit from TKI therapy.
- Published
- 2016
45. Conventional and Advanced Imaging of Diffuse Intrinsic Pontine Glioma
- Author
-
Robert J. Young, Kyung K. Peck, Sofia Haque, and Jamie Tisnado
- Subjects
Mri techniques ,medicine.medical_specialty ,business.industry ,Brain Stem Neoplasm ,Neuroimaging ,Glioma ,medicine.disease ,Radiation planning ,Article ,03 medical and health sciences ,0302 clinical medicine ,Pediatric brain ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,medicine ,Brain Stem Neoplasms ,Humans ,Neurology (clinical) ,Radiology ,Child ,business ,030217 neurology & neurosurgery ,Brain Stem - Abstract
Diffuse intrinsic pontine glioma is the most common brainstem tumor in pediatric patients. This tumor remains one of the most deadly pediatric brain tumors. The diagnosis primarily relies on clinical symptoms and imaging findings. Conventional MRI provides a noninvasive accurate method of diagnosis of these tumors. Advanced MRI techniques are becoming more widely used and studied as additional noninvasive methods to assist clinicians in initial diagnosis and staging, monitoring disease, as well as in surgical and radiation planning. This article will provide an overview of DIPG and describe the typical imaging findings with a focus on advanced imaging techniques.
- Published
- 2016
46. Second-opinion interpretations of neuroimaging studies by oncologic neuroradiologists can help reduce errors in cancer care
- Author
-
Jung Hun Oh, Vaios Hatzoglou, Ian Ganly, Andrei I. Holodny, Antonio Omuro, Joshua Gaal, Amita Shukla-Dave, Robin Fatovic, Yasmin Khakoo, and Sofia Haque
- Subjects
Cancer Research ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Second opinion ,Cancer ,Retrospective cohort study ,Magnetic resonance imaging ,Disease ,medicine.disease ,030218 nuclear medicine & medical imaging ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Neuroimaging ,030220 oncology & carcinogenesis ,medicine ,Medical physics ,Stage (cooking) ,Young adult ,business - Abstract
BACKGROUND The purpose of this study was to investigate the utility and clinical impact of second-opinion interpretations of outside neuroimaging studies by oncologic neuroradiologists at a National Cancer Institute–designated cancer center. METHODS We performed a retrospective analysis of initial outside and second-opinion radiology reports from 300 computed tomography and magnetic resonance imaging studies and identified cases with discrepancies between the two reports. An adult neuro-oncologist, pediatric neuro-oncologist, and head and neck surgeon reviewed each pair of discrepant reports based on their area of expertise, patient age, and the type of study performed. The clinicians were blinded to the origin of each report and recorded whether the differences in the reports would have led to a change in patient management and/or disease staging. Histopathologic analysis, clinical assessment, and/or minimum 3-month imaging follow-up served as the reference standards to establish which of the 2 reports was correct. RESULTS Among the 283 cases that met our study criteria, there were 55 neuroimaging studies with disagreements (19%) between the initial outside report and second-opinion interpretation. Patient management and/or disease stage would have been altered in 42 of 283 cases (15%) based on report differences as determined by the 2 neuro-oncologists and the surgeon participating in the study. Sufficient follow-up was available in 35 of 42 cases (83%). The second-opinion interpretation was correct 100% of the time (35/35). CONCLUSION Second-opinion interpretations of neuroimaging studies by subspecialized oncologic neuroradiologists provide added value by reducing error and optimizing the care of cancer patients. Cancer 2016. © 2016 American Cancer Society. Cancer 2016;122:2708–2714. © 2016 American Cancer Society.
- Published
- 2016
47. Abstract P4-14-21: A phase I trial of ganetespib (heat shock protein 90 inhibitor) in combination with paclitaxel and trastuzumab in patients with human epidermal growth factor receptor-2 positive (HER2+) metastatic breast cancer (MBC)
- Author
-
S. Patil, S Chandarlapaty, Francisco J. Esteva, Karen Cadoo, Sofia Haque, Deirdre Neville, David B. Solit, Shanu Modi, Sylvia Adams, Clifford A. Hudis, Kent Friedman, James L. Speyer, Komal Jhaveri, Eleonora Teplinsky, and Gabriella D'Andrea
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Ganetespib ,Pharmacology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Trastuzumab ,medicine ,skin and connective tissue diseases ,neoplasms ,Triple-negative breast cancer ,Chemotherapy ,business.industry ,medicine.disease ,Metastatic breast cancer ,030104 developmental biology ,Oncology ,Docetaxel ,Paclitaxel ,chemistry ,030220 oncology & carcinogenesis ,Pertuzumab ,business ,medicine.drug - Abstract
Introduction: Targeted therapies in HER2+ MBC significantly improve outcomes but efficacy is limited by therapeutic resistance. HSP90 is a molecular chaperone involved in the stability and function of multiple signaling onco-proteins. HER2 is an acutely sensitive HSP90 client and HSP90 inhibition can overcome trastuzumab resistance. Our group reported objective responses with 17-AAG plus trastuzumab in HER2+ MBC. Ganetespib, a synthetic, second generation HSP90 inhibitor has increased potency and tolerability compared with earlier agents. We reported anti-tumor activity in metastatic HER2+ and triple negative breast cancer with single agent ganetespib. Preclinically, HSP90 inhibition has synergistic anti-tumor activity with taxanes and trastuzumab. This study will define the MTD and RP2D of ganetespib plus paclitaxel and trastuzumab in HER2+ MBC. Methods: In this 3+3 phase I dose escalation study, patients with trastuzumab-resistant HER2+ MBC receive weekly trastuzumab and paclitaxel (80mg/m2) with ganetespib on day 1, 8, 15 of a 28 day cycle. HR+ positive patients are required to have at least one prior line of endocrine therapy. DLT of ganetespib monotherapy is diarrhea and therefore patients receive prophylactic anti-motility agents. Based on prior experience with ganetespib plus docetaxel in NSCLC, only 3 dose levels of ganetespib were explored: 100mg/m2, 150mg/m2 and a 3rd cohort of 125mg/m2, if needed. Secondary endpoints include evaluation of effects of ganetespib on the pharmacokinetics (PK) of paclitaxel and preliminary efficacy assessment. Results: The dosing cohorts (100 mg/m2 (n=3) and 150 mg/m2 (n=6)) have been completed without any DLTs. Median age was 46 years (range 29-65), median prior lines of chemotherapy and anti-HER2 therapy were 3 (range 2-6) and 3 (range 2-4) respectively, including prior pertuzumab in 9/9 and T-DM1 in 8/9 patients. There were no grade 3/4 adverse events (AEs) related to ganetespib. Most common AEs related to ganetespib were diarrhea, fatigue, anemia and rash. Paclitaxel PK data available from 6/9 patients are not appreciably different from those reported in literature. Overall response rate was 25% (2/8 had PR in 150 mg/m2 cohort; 1 patient was not evaluable), SD in 63% (5/8), and clinical benefit rate (CR+PR+SD>24 weeks) was 50% (4/8). 3 patients remain on study. Conclusion: The RP2D of ganetespib is 150mg/m2 in combination with paclitaxel and trastuzumab. The combination was safe and well tolerated. Updated PFS and PK data will be presented. Despite prior taxanes, pertuzumab and T-DM1, clinical activity of this triplet regimen in this heavily pre-treated cohort is very promising and together with our prior experience with 17-AAG plus trastuzumab and single agent ganetespib warrants further study in HER2+ MBC. A phase 2 trial is being planned in trastuzumab-refractory HER2+ MBC who have progressed on prior pertuzumab and T-DM1. Additionally, the protocol is amended to assess the safety of ganetespib in combination with paclitaxel, trastuzumab and pertuzumab in the first-line setting. Citation Format: Jhaveri K, Teplinsky E, Chandarlapaty S, Solit D, Cadoo K, Speyer J, D'Andrea G, Adams S, Patil S, Haque S, Friedman K, Neville D, Esteva F, Hudis C, Modi S. A phase I trial of ganetespib (heat shock protein 90 inhibitor) in combination with paclitaxel and trastuzumab in patients with human epidermal growth factor receptor-2 positive (HER2+) metastatic breast cancer (MBC). [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr P4-14-21.
- Published
- 2016
48. Unilateral Retinoblastoma Metastatic to the Skull and Both Orbits
- Author
-
Jasmine H. Francis, David H. Abramson, and Sofia Haque
- Subjects
medicine.medical_specialty ,business.industry ,Retinal Neoplasms ,Skull ,Retinoblastoma ,Magnetic Resonance Imaging ,Skull Base Neoplasms ,Ophthalmology ,medicine.anatomical_structure ,Child, Preschool ,Humans ,Orbital Neoplasms ,Medicine ,Female ,Neoplasm Metastasis ,business ,Orbit ,Unilateral Retinoblastoma - Published
- 2020
49. A phase I/Ib study of lenvatinib and cetuximab in patients with recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC)
- Author
-
Loren S. Michel, Kenneth K.-S. Ng, Alan Loh Ho, Juliana Eng, Wanqing Iris Zhi, Sofia Haque, David G. Pfister, Lara Dunn, Anuja Kriplani, Elizabeth Warner, James Vincent Fetten, and Eric J. Sherman
- Subjects
Cancer Research ,Cetuximab ,business.industry ,medicine.disease ,Head and neck squamous-cell carcinoma ,stomatognathic diseases ,chemistry.chemical_compound ,Oncology ,chemistry ,Fibroblast growth factor receptor ,otorhinolaryngologic diseases ,Cancer research ,medicine ,In patient ,Lenvatinib ,business ,neoplasms ,medicine.drug - Abstract
6541 Background: Despite overexpression of EGFR in HNSCC, cetuximab monotherapy has limited benefit. Fibroblast growth factor receptor (FGFR) signaling is a known resistance mechanism to EGFR inhibition. Lenvatinib is a multi-targeted receptor tyrosine kinase inhibitor (RTKI) and has unique activity against FGFR 1,2,3, and 4. We are evaluating inhibition of EGFR and RTKs including FGFR through the combination of cetuximab and lenvatinib in patients (pts) with R/M HNSCC. Methods: In this phase I/Ib, single-institution study, pts with measurable disease per RECIST v1.1 that is incurable with surgery and radiation are eligible regardless of prior cetuximab therapy. The dose de-escalation phase included pts with HNSCC and cutaneous squamous cell carcinoma (cSCC) treated with standard cetuximab dosing and lenvatinib in 3 potential dose levels (DL): (0) 24mg, (-1) 20mg, (-2) 14mg oral daily in a standard 3+3 design. The primary objective was to determine the MTD of lenvatinib in combination with cetuximab. The expansion phase included an additional 5 pts with HNSCC treated at the MTD. Exploratory endpoints include ORR and PFS in HNSCC pts treated at the MTD. Results: 12 evaluable pts were treated on the dose de-escalation phase. There were no DLTs on DL 0; however, 3/6 pts were removed immediately following the 28-day DLT period due to toxicity that included extensive thrombotic events and athlerosclerotic disease. On DL -1, 0/6 pts (5 HNSCC/1 cSCC) had a DLT establishing lenvatinib 20mg daily as the MTD. 7 pts were enrolled onto the expansion phase; 4 are currently evaluable for response and 2 are unevaluable because of withdrawal due to a cetuximab reaction and required surgery. Of the 9 evaluable HNSCC pts treated with lenvatinib 20mg daily, 6 pts had a PR with a 67% ORR. For the 8 pts who have completed treatment, the median PFS is 3.6 months (range 1.6-10.4). Grade 3 AEs regardless of attribution included hypertension (3), oral mucositis (3) and oral cavity fistula (1). The most common AEs were acneiform rash (7), fatigue (6), and hypertension/hypothyroidism/oral mucositis (5 each). Conclusions: The MTD of lenvatinib 20mg daily with cetuximab appears to be active in R/M HSNCC with an impressive preliminary ORR, warranting further evaluation of the efficacy of this combination. Clinical trial information: NCT03524326 .
- Published
- 2020
50. Radioiodine (RAI) in combination with durvalumab for recurrent/metastatic thyroid cancers
- Author
-
Loren S. Michel, Alan Loh Ho, Eric J. Sherman, Jeffrey A. Knauf, Irina Ostrovnaya, Lara Dunn, R. Michael Tuttle, Ravinder K. Grewal, Ronald Ghossein, Elizabeth Warner, Bharat Burman, Sofia Haque, James A. Fagin, Anuja Kriplani, Stephanie Fish, James Vincent Fetten, Mona M. Sabra, Laura Boucai, and David G. Pfister
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Durvalumab ,business.industry ,Thyroid ,Autoimmune thyroid disease ,medicine.disease ,Immune checkpoint ,Blockade ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,High incidence ,business ,Thyroid cancer ,030215 immunology - Abstract
6587 Background: Immune checkpoint blockade (ICB) has limited efficacy for radioiodine-refractory thyroid cancer. The high incidence of autoimmune thyroid disease and ICB-induced hypothyroidism suggests that loss of T cell tolerance to thyroid protein epitopes is common and can be activated by ICB to induce immune responses. We hypothesize that RAI can enhance presentation of thyroid protein immunogens and putative neoantigens in thyroid cancers to amplify the effectiveness of ICB. We studied the safety and efficacy of RAI plus the anti-PD-L1 agent durvalumab (durva) in recurrent/metastatic (R/M) patients (pts). Methods: Pts. had at least one RAI-avid tumor on the most recent RAI scan or one tumor on FDG PET with an SUVmax < 10. RECIST measurable disease was required. Any number of prior therapies was allowed. Pts were treated with durva 1500 mg IV every 4 weeks with recombinant human TSH (rhTSH)-stimulated RAI (100 mCi) administered in Cycle 1. Treatment beyond progression was allowed. The primary objective was to assess safety. Durva related dose limiting toxicities (DLTs) were monitored for 6 weeks after the first dose. Since no durva DLTs were observed in the first 6 pts, per protocol rules the trial accrued 11 pts total. Secondary objectives were assessing best overall response (BOR) per RECIST and progression-free survival (PFS). Results: 11 pts (7 female) were enrolled. Eight had prior drug therapy. No DLTs or > Grade 3 durva related adverse events (AEs) were observed. The most common non-laboratory AEs (regardless of attribution) were cough (7), hypertension (7), pain (6), edema (5), and fatigue/nausea/diarrhea/arthralgia/dry skin/dyspnea/edema (4 each). As of 2/6/20, 2 had partial response, 7 stable disease, and 2 progression of disease as BOR. Six pts had tumor regression. Four pts received treatment for > 6 months. Six are still on treatment. Analyses of research biopsies (bxs) (8 had pre-treatment bxs, 6 had an additional on-treatment bx) will be presented. Conclusions: Durva plus RAI is safe and well tolerated. The preliminary efficacy signal in this small cohort is promising. Understanding how RAI plus PD-L1 targeting impacts the tumor immune microenvironment may guide how RAI should be evaluated in future ICB trials. Clinical trial information: NCT03215095 .
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.