Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
AJR Am J Roentgenol ; 221(6): 760-772, 2023 12.
Article in English | MEDLINE | ID: mdl-37436033

ABSTRACT

BACKGROUND. Imaging reports that consistently document all disease sites with a potential to increase surgical complexity or morbidity can facilitate ovarian cancer treatment planning. OBJECTIVE. The aims of this study were to compare simple structured reports and synoptic reports from pretreatment CT examinations in patients with advanced ovarian cancer in terms of completeness of documenting involvement of clinically relevant anatomic sites as well as to evaluate physician satisfaction with synoptic reports. METHODS. This retrospective study included 205 patients (median age, 65 years) who underwent contrast-enhanced abdominopelvic CT before primary treatment of advanced ovarian cancer from June 1, 2018, to January 31, 2022. A total of 128 reports generated on or before March 31, 2020, used a simple structured report (free text organized into sections); 77 reports generated on or after April 1, 2020, used a synoptic report (a list of 45 anatomic sites relevant to ovarian cancer management, each of which was classified in terms of disease absence versus presence). Reports were reviewed for completeness of documentation of involvement of the 45 sites. For patients who underwent neoadjuvant chemotherapy based on diagnostic laparoscopy findings or underwent primary debulking surgery with suboptimal resection, the EMR was reviewed to identify surgically established sites of disease that were unresectable or challenging to resect. Gynecologic oncology surgeons were electronically surveyed. RESULTS. The mean report turnaround time was 29.8 minutes for simple structured reports versus 54.5 minutes for synoptic reports (p < .001). A mean of 17.6 of 45 sites (range, four to 43 sites) were mentioned by simple structured reports versus 44.5 of 45 sites (range, 39-45) for synoptic reports (p < .001). Forty-three patients had surgically established unresectable or challenging-to-resect disease; involvement of anatomic site(s) with such disease was mentioned in 37% (11/30) of simple structured reports versus 100% (13/13) of synoptic reports (p < .001). All eight surveyed gynecologic oncology surgeons completed the survey. CONCLUSION. A synoptic report improved completeness of pretreatment CT reports in patients with advanced ovarian cancer, including for established sites of unresectable or challenging-to-resect disease. CLINICAL IMPACT. The findings indicate the role of disease-specific synoptic reports in facilitating referrer communication and potentially guiding clinical decision-making.


Subject(s)
Genital Neoplasms, Female , Ovarian Neoplasms , Physicians , Humans , Female , Aged , Retrospective Studies , Patient Satisfaction , Ovarian Neoplasms/diagnostic imaging , Ovarian Neoplasms/surgery , Documentation , Tomography, X-Ray Computed , Personal Satisfaction
2.
Cancer ; 126(6): 1274-1282, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31880826

ABSTRACT

BACKGROUND: PI3K pathway activation is common in endometrial cancer. We evaluated the safety and efficacy of the dual PI3K/mTOR inhibitor, LY3023414, in patients with advanced endometrial cancer harboring activating mutations in the PI3K pathway. METHODS: We conducted a single-arm phase 2 study of monotherapy LY3023414. Eligible patients had advanced endometrial cancer of any grade, prior management with 1-4 cytotoxic lines, and PI3K pathway activation prospectively defined as a loss-of-function PTEN alteration or activating alteration in PIK3CA, AKT1, PIK3R1, PIK3R2, or MTOR. The primary objective was best overall response rate (ORR) per RECIST 1.1. RESULTS: Twenty-eight patients were treated; histologies included endometroid (39%), carcinosarcoma (25%), serous (21%), and mixed (14%). Patients were heavily pretreated, with a median of 2 prior cytotoxic lines (range, 1-3). The most common alterations involved PIK3CA (68%), PTEN (43%), and PIK3R1 (32%). In the 25 efficacy-evaluable patients, the ORR was 16% (90% CI, 7%-100%), and the clinical benefit rate was 28% (90% CI, 16%-100%). Four patients had a confirmed partial response, and 2 responses lasted for >9 months. The median progression-free survival and overall survival were 2.5 months (95% CI, 1.2-3.0) and 9.2 months (95% CI, 5.0-15.9), respectively. The most common all-grade treatment-related adverse events were anemia (71%), hyperglycemia (71%), hypoalbuminemia (68%), and hypophosphatemia (61%). No correlation between molecular alterations and response was observed. CONCLUSION: In patients with heavily pretreated advanced endometrial cancer prospectively selected for tumors with activating PI3K pathway mutations, LY3023414 demonstrated modest single-agent activity and a manageable safety profile.


Subject(s)
Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Mutation , Phosphatidylinositol 3-Kinases/genetics , Pyridines/therapeutic use , Quinolones/therapeutic use , Aged , Class I Phosphatidylinositol 3-Kinases , Class Ia Phosphatidylinositol 3-Kinase/genetics , Endometrial Neoplasms/pathology , Enzyme Activation , Female , Humans , Hyperglycemia/chemically induced , Hypoalbuminemia/chemically induced , Hypophosphatemia/chemically induced , Middle Aged , PTEN Phosphohydrolase/genetics , Progression-Free Survival , Proto-Oncogene Proteins c-akt/genetics , Pyridines/adverse effects , Quinolones/adverse effects , Signal Transduction , TOR Serine-Threonine Kinases , Treatment Outcome
3.
Gynecol Oncol ; 158(3): 608-613, 2020 09.
Article in English | MEDLINE | ID: mdl-32518012

ABSTRACT

OBJECTIVE: To describe outcomes using a multimodal algorithm to triage patients with advanced epithelial ovarian cancer (EOC) to primary debulking surgery (PDS) versus neoadjuvant chemotherapy (NACT). METHODS: All patients with EOC treated at our institution from 04/2015-08/2018 were identified. We included patients without contraindication to PDS who underwent prospective calculation of a Resectability (R)-score. A low risk score for suboptimal cytoreduction was defined as ≤6, and a high risk score ≥7. Patients were triaged to laparotomy/PDS, laparoscopic evaluation of resectability (LSC), or NACT depending on R-score. RESULTS: Among 299 participants, 226 (76%) had a low risk score and 73 (24%) a high risk score. For patients with a low risk score, management included laparotomy/PDS, 181 (80%); LSC, 43 (19%) (with subsequent triage: PDS, 31; NACT, 12); and NACT, 2 (1%). For patients with a high risk score, management included laparotomy/PDS, 9 (12%); LSC, 51 (70%) (with subsequent triage: PDS, 28; NACT, 23); and NACT, 13 (18%). Overall, 83% underwent PDS, with a 75% CGR rate and 94% optimal cytoreduction rate. Use of the algorithm resulted in a 31% LSC rate and a 6% rate of suboptimal PDS. CONCLUSIONS: The multimodal algorithm led to excellent surgical results; 94% of patients achieved an optimal resection, with a very low rate of suboptimal cytoreduction.


Subject(s)
Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/surgery , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/surgery , Triage/methods , Adult , Aged , Aged, 80 and over , Algorithms , Carcinoma, Ovarian Epithelial/pathology , Cytoreduction Surgical Procedures/methods , Female , Humans , Middle Aged , Neoadjuvant Therapy , Ovarian Neoplasms/pathology , Treatment Outcome
4.
Clin Cancer Res ; 30(17): 3812-3823, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38922339

ABSTRACT

PURPOSE: Even though BRAF fusions are increasingly detected in standard multigene next-generation sequencing panels, few reports have explored their structure and impact on clinical course. EXPERIMENTAL DESIGN: We collected data from patients with BRAF fusion-positive cancers identified through a genotyping protocol of 97,024 samples. Fusions were characterized and reviewed for oncogenic potential (in-frame status, non-BRAF partner gene, and intact BRAF kinase domain). RESULTS: We found 241 BRAF fusion-positive tumors from 212 patients with 82 unique 5' fusion partners spanning 52 histologies. Thirty-nine fusion partners were not previously reported, and 61 were identified once. BRAF fusion incidence was enriched in pilocytic astrocytomas, gangliogliomas, low-grade neuroepithelial tumors, and acinar cell carcinoma of the pancreas. Twenty-four patients spanning multiple histologies were treated with MAPK-directed therapies, of which 20 were evaluable for RECIST. Best response was partial response (N = 2), stable disease (N = 11), and progressive disease (N = 7). The median time on therapy was 1 month with MEK plus BRAF inhibitors [(N = 11), range 0-18 months] and 8 months for MEK inhibitors [(N = 14), range 1-26 months]. Nine patients remained on treatment for longer than 6 months [pilocytic astrocytomas (N = 6), Erdheim-Chester disease (N = 1), extraventricular neurocytoma (N = 1), and melanoma (N = 1)]. Fifteen patients had acquired BRAF fusions. CONCLUSIONS: BRAF fusions are found across histologies and represent an emerging actionable target. BRAF fusions have a diverse set of fusion partners. Durable responses to MAPK therapies were seen, particularly in pilocytic astrocytomas. Acquired BRAF fusions were identified after targeted therapy, underscoring the importance of postprogression biopsies to optimize treatment at relapse in these patients.


Subject(s)
Oncogene Proteins, Fusion , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Adult , Male , Middle Aged , Female , Aged , Oncogene Proteins, Fusion/genetics , Young Adult , Adolescent , Molecular Targeted Therapy , Child , Neoplasms/genetics , Neoplasms/pathology , Biomarkers, Tumor/genetics , Genomics/methods , Child, Preschool , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , High-Throughput Nucleotide Sequencing
5.
Cancer Discov ; 12(5): 1233-1247, 2022 05 02.
Article in English | MEDLINE | ID: mdl-35135829

ABSTRACT

NRG1 rearrangements are recurrent oncogenic drivers in solid tumors. NRG1 binds to HER3, leading to heterodimerization with other HER/ERBB kinases, increased downstream signaling, and tumorigenesis. Targeting ERBBs, therefore, represents a therapeutic strategy for these cancers. We investigated zenocutuzumab (Zeno; MCLA-128), an antibody-dependent cellular cytotoxicity-enhanced anti-HER2xHER3 bispecific antibody, in NRG1 fusion-positive isogenic and patient-derived cell lines and xenograft models. Zeno inhibited HER3 and AKT phosphorylation, induced expression of apoptosis markers, and inhibited growth. Three patients with chemotherapy-resistant NRG1 fusion-positive metastatic cancer were treated with Zeno. Two patients with ATP1B1-NRG1-positive pancreatic cancer achieved rapid symptomatic, biomarker, and radiographic responses and remained on treatment for over 12 months. A patient with CD74-NRG1-positive non-small cell lung cancer who had progressed on six prior lines of systemic therapy, including afatinib, responded rapidly to treatment with a partial response. Targeting HER2 and HER3 simultaneously with Zeno is a novel therapeutic paradigm for patients with NRG1 fusion-positive cancers. SIGNIFICANCE: NRG1 rearrangements encode chimeric ligands that activate the ERBB receptor tyrosine kinase family. Here we show that targeting HER2 and HER3 simultaneously with the bispecific antibody Zeno leads to durable clinical responses in patients with NRG1 fusion-positive cancers and is thus an effective therapeutic strategy. This article is highlighted in the In This Issue feature, p. 1171.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Antibodies, Bispecific , Carcinogenesis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Gene Rearrangement , Humans , Immunoglobulin G , Lung Neoplasms/genetics , Neuregulin-1/genetics , Receptor, ErbB-2 , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism
6.
Radiology ; 258(1): 41-56, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21183492

ABSTRACT

Cancer chemotherapy has evolved from cytotoxic agents and now includes several new agents that target specific molecules responsible for the regulation of cell growth, nutrient supply, and differentiation. These molecularly targeted therapies have a different mechanism of action than do classic cytotoxic agents, which predominantly attack rapidly proliferating cells. Not surprisingly, therefore, the toxicity of targeted and cytotoxic agents may differ in both clinical and radiologic presentation. Many of the toxicities of targeted therapies are not cumulative or dose dependent, some are asymptomatic, and others may first manifest radiologically. It is imperative that radiologists be aware of these toxicities and that they learn to recognize the relevant findings so that they can provide a complete differential diagnosis and thus play an important role in patient care.


Subject(s)
Antineoplastic Agents/adverse effects , Molecular Targeted Therapy/adverse effects , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Tomography, X-Ray Computed , Female , Female Urogenital Diseases/chemically induced , Female Urogenital Diseases/diagnostic imaging , Gastrointestinal Diseases/chemically induced , Gastrointestinal Diseases/diagnostic imaging , Humans , Liver Diseases/diagnostic imaging , Liver Diseases/etiology , Lung Diseases/chemically induced , Lung Diseases/diagnostic imaging , Male , Male Urogenital Diseases/chemically induced , Male Urogenital Diseases/diagnostic imaging , Pancreatic Diseases/chemically induced , Pancreatic Diseases/diagnostic imaging , Vascular Diseases/chemically induced , Vascular Diseases/diagnostic imaging
7.
Clin Imaging ; 71: 29-33, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33171363

ABSTRACT

OBJECTIVE: mTOR inhibitor-associated pneumonitis is common and often asymptomatic. We describe a waxing and waning pattern of pneumonitis observed on computed tomography (CT) scans of patients with renal cell carcinoma who were being treated with mTOR inhibitor molecular targeted therapy. MATERIALS AND METHODS: In this HIPAA-compliant, IRB-approved retrospective single-institution study, 25 renal cell carcinoma patients were identified who received single-therapy temsirolimus or everolimus between January 2011 and June 2015 and who had chest CT scans available for review both before and after initiation of mTOR inhibitor treatment. A detailed review of the electronic medical record and serial chest CT examinations was performed. RESULTS: Radiologic findings compatible with pneumonitis were identified in 13/25 (52%) patients on mTOR inhibitors in our study. Of the patients with CT findings of pneumonitis, 8/13 (62%) demonstrated a waxing and waning pattern; of whom 7 had clinical symptoms of pneumonitis. Of the 17 patients who received temsirolimus, 9/17 (53%) developed radiologic findings compatible with pneumonitis and 4/9 (44%) developed a waxing and waning pattern. Of the 8 patients who received everolimus, 4/8 (50%) had radiologic findings compatible with pneumonitis and 4/4 (100%) developed a waxing and waning pattern. CONCLUSION: Waxing and waning is an unrecognized pattern of mTOR inhibitor-associated pneumonitis. Recognition of this pattern will promote clinical-radiologic concordance and may facilitate patient management.


Subject(s)
Antineoplastic Agents , Carcinoma, Renal Cell , Kidney Neoplasms , Pneumonia , Antineoplastic Agents/adverse effects , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/drug therapy , Humans , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/drug therapy , Pneumonia/chemically induced , Pneumonia/diagnostic imaging , Retrospective Studies , TOR Serine-Threonine Kinases/therapeutic use
8.
Clin Cancer Res ; 27(1): 96-106, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33046513

ABSTRACT

PURPOSE: Intratumorally injected Clostridium novyi-NT (nontoxic; lacking the alpha toxin), an attenuated strain of C. novyi, replicates within hypoxic tumor regions resulting in tumor-confined cell lysis and inflammatory response in animals, which warrants clinical investigation. PATIENTS AND METHODS: This first-in-human study (NCT01924689) enrolled patients with injectable, treatment-refractory solid tumors to receive a single intratumoral injection of C. novyi-NT across 6 dose cohorts (1 × 104 to 3 × 106 spores, 3+3 dose-escalation design) to determine dose-limiting toxicities (DLT), and the maximum tolerated dose. RESULTS: Among 24 patients, a single intratumoral injection of C. novyi-NT led to bacterial spores germination and the resultant lysis of injected tumor masses in 10 patients (42%) across all doses. The cohort 5 dose (1 × 106 spores) was defined as the maximum tolerated dose; DLTs were grade 4 sepsis (n = 2) and grade 4 gas gangrene (n = 1), all occurring in three patients with injected tumors >8 cm. Other treatment-related grade ≥3 toxicities included pathologic fracture (n = 1), limb abscess (n = 1), soft-tissue infection (n = 1), respiratory insufficiency (n = 1), and rash (n = 1), which occurred across four patients. Of 22 evaluable patients, nine (41%) had a decrease in size of the injected tumor and 19 (86%) had stable disease as the best overall response in injected and noninjected lesions combined. C. novyi-NT injection elicited a transient systemic cytokine response and enhanced systemic tumor-specific T-cell responses. CONCLUSIONS: Single intratumoral injection of C. novyi-NT is feasible. Toxicities can be significant but manageable. Signals of antitumor activity and the host immune response support additional studies of C. novyi-NT in humans.


Subject(s)
Clostridium/immunology , Immunotherapy/methods , Neoplasms/therapy , Spores, Bacterial/immunology , Adult , Aged , Drug Resistance, Neoplasm/immunology , Feasibility Studies , Female , Humans , Immunotherapy/adverse effects , Injections, Intralesional , Male , Middle Aged , Neoplasms/immunology
9.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32503949

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) to date have demonstrated limited activity in advanced ovarian cancer (OC). Folate receptor alpha (FRα) is overexpressed in the majority of OCs and presents an attractive target for a combination immunotherapy to potentially overcome resistance to ICI in OCs. The current study sought to examine clinical and immunologic responses to TPIV200, a multiepitope FRα vaccine administered with programmed death ligand 1 (PD-L1) inhibitor durvalumab in patients with advanced platinum-resistant OC. METHODS: Following Simon two-stage phase II trial design, 27 patients were enrolled. Treatment was administered in 28-day cycles (intradermal TPIV200 and granulocyte-macrophage colony-stimulating factor (GM-CSF) for 6 cycles and intravenous durvalumab for 12 cycles). Primary endpoints included overall response rate and progression-free survival at 24 weeks. Translational parameters focused on tumor microenvironment, PD-L1 and FRα expression, and peripheral vaccine-specific immune responses. RESULTS: Treatment was well tolerated, with related grade 3 toxicity rate of 18.5%. Increased T cell responses to the majority of peptides were observed in all patients at 6 weeks (p<0.0001). There was one unconfirmed partial response (3.7%) and nine patients had stable disease (33.3%). Clinical benefit was not associated with baseline FRα or PD-L1 expression. One patient with prolonged clinical benefit demonstrated loss of FRα expression and upregulation of PD-L1 in a progressing lesion. Despite the low overall response rate, the median overall survival was 21 months (13.5-∞), with evidence of benefit from postimmunotherapy regimens. CONCLUSIONS: Combination of TPIV200 and durvalumab was safe and elicited robust FRα-specific T cell responses in all patients. Unexpectedly durable survival in this heavily pretreated population highlights the need to investigate the impact of FRα vaccination on the OC biology post-treatment.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Biomarkers, Tumor/immunology , Cancer Vaccines/therapeutic use , Folate Receptor 1/immunology , Ovarian Neoplasms/drug therapy , Tumor Microenvironment/immunology , Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma, Clear Cell/immunology , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Antineoplastic Agents, Immunological/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Drug Therapy, Combination , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Non-Randomized Controlled Trials as Topic , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Prognosis , Survival Rate , Treatment Outcome
10.
JAMA Oncol ; 4(3): 384-388, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29188284

ABSTRACT

IMPORTANCE: The histiocytic neoplasms Erdheim-Chester disease (ECD) and Langerhans cell histiocytosis (LCH) are highly enriched for BRAF V600 mutations and have been previously shown to be responsive to treatment with vemurafenib, an inhibitor of the BRAF V600 kinase. However, the long-term efficacy and safety of prolonged vemurafenib use in these patients are not defined. Here we analyze the final efficacy and safety data for vemurafenib in patients with ECD and LCH enrolled in the VE-BASKET study. OBJECTIVE: To determine the efficacy and safety of vemurafenib in adults with ECD or LCH enrolled in the VE-BASKET study. DESIGN, SETTING, AND PARTICIPANTS: The VE-BASKET study was an open-label, nonrandomized, multicohort study for patients with nonmelanoma cancers harboring the BRAF V600 mutation. Patients with BRAF V600-mutant ECD or LCH were enrolled in an "other solid tumor" cohort of the VE-BASKET study, and they were enrolled in the present study. INTERVENTIONS: Patients received vemurafenib, 960 mg, twice daily continuously until disease progression, study withdrawal, or occurrence of intolerable adverse effects. MAIN OUTCOMES AND MEASURES: The primary end point was confirmed objective response rate (ORR) by Response Evaluation Criteria in Solid Tumors (RECIST, version 1.1). Secondary end points included progression-free survival (PFS), overall survival (OS), metabolic response by modified positron-emission tomography (PET) Response Criteria in Solid Tumors (PERCIST) using 18F-fluorodeoxyglucose (FDG)-PET/computed tomography (CT), and safety. RESULTS: A total of 26 patients from the VE-BASKET trial (22 with ECD, 4 with LCH) were included in the present study (14 women and 12 men; median age, 61 years; age range, 51-74 years). The confirmed ORR was 61.5% (95% CI, 40.6%-79.8%) in the overall cohort and 54.5% (95% CI, 32.2%-75.6%) in patients with ECD. All evaluable patients achieved stable disease or better. The median PFS and OS had not been reached in the overall cohort at study closure despite a median follow-up of 28.8 months; 2-year PFS was 86% (95% CI, 72%-100%), and 2-year OS was 96% (95% CI, 87%-100%). All 15 patients evaluated by FDG-PET/CT achieved a metabolic response, including 12 patients (80%) with a complete metabolic response. The most common adverse events (AEs) in the overall cohort included arthralgia, maculopapular rash, fatigue, alopecia, prolonged QT interval, skin papilloma, and hyperkeratosis. Hypertension and dermatologic AEs occurred at higher rates than those reported in metastatic melanoma. CONCLUSIONS AND RELEVANCE: In this study, vemurafenib had prolonged efficacy in patients with BRAF V600-mutant ECD and LCH and warrants consideration as a new standard of care for these patients.


Subject(s)
Erdheim-Chester Disease/drug therapy , Erdheim-Chester Disease/genetics , Histiocytosis, Langerhans-Cell/drug therapy , Mutation, Missense , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/therapeutic use , Aged , Amino Acid Substitution/genetics , Erdheim-Chester Disease/diagnosis , Erdheim-Chester Disease/mortality , Female , Fluorodeoxyglucose F18 , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/mortality , Humans , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Prognosis , Survival Analysis , Treatment Outcome , Valine/genetics
11.
J Clin Oncol ; 35(20): 2251-2259, 2017 Jul 10.
Article in English | MEDLINE | ID: mdl-28489509

ABSTRACT

Purpose AKT1 E17K mutations are oncogenic and occur in many cancers at a low prevalence. We performed a multihistology basket study of AZD5363, an ATP-competitive pan-AKT kinase inhibitor, to determine the preliminary activity of AKT inhibition in AKT-mutant cancers. Patients and Methods Fifty-eight patients with advanced solid tumors were treated. The primary end point was safety; secondary end points were progression-free survival (PFS) and response according to Response Evaluation Criteria in Solid Tumors (RECIST). Tumor biopsies and plasma cell-free DNA (cfDNA) were collected in the majority of patients to identify predictive biomarkers of response. Results In patients with AKT1 E17K-mutant tumors (n = 52) and a median of five lines of prior therapy, the median PFS was 5.5 months (95% CI, 2.9 to 6.9 months), 6.6 months (95% CI, 1.5 to 8.3 months), and 4.2 months (95% CI, 2.1 to 12.8 months) in patients with estrogen receptor-positive breast, gynecologic, and other solid tumors, respectively. In an exploratory biomarker analysis, imbalance of the AKT1 E17K-mutant allele, most frequently caused by copy-neutral loss-of-heterozygosity targeting the wild-type allele, was associated with longer PFS (hazard ratio [HR], 0.41; P = .04), as was the presence of coincident PI3K pathway hotspot mutations (HR, 0.21; P = .045). Persistent declines in AKT1 E17K in cfDNA were associated with improved PFS (HR, 0.18; P = .004) and response ( P = .025). Responses were not restricted to patients with detectable AKT1 E17K in pretreatment cfDNA. The most common grade ≥ 3 adverse events were hyperglycemia (24%), diarrhea (17%), and rash (15.5%). Conclusion This study provides the first clinical data that AKT1 E17K is a therapeutic target in human cancer. The genomic context of the AKT1 E17K mutation further conditioned response to AZD5363.


Subject(s)
Antineoplastic Agents/adverse effects , DNA, Neoplasm/blood , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Pyrimidines/adverse effects , Pyrroles/adverse effects , Adult , Aged , Alleles , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Diarrhea/chemically induced , Disease-Free Survival , Drug Eruptions/etiology , Exanthema/chemically induced , Female , Humans , Hyperglycemia/chemically induced , Loss of Heterozygosity , Male , Middle Aged , Neoplasms/blood , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Response Evaluation Criteria in Solid Tumors , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL