Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lipika Goyal is active.

Publication


Featured researches published by Lipika Goyal.


Annals of Surgery | 2015

Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer.

Cristina R. Ferrone; Giovanni Marchegiani; Theodore S. Hong; David P. Ryan; Vikram Deshpande; Erin McDonnell; Francesco Sabbatino; Daniela Dias Santos; Jill N. Allen; Lawrence S. Blaszkowsky; Jeffrey W. Clark; Jason E. Faris; Lipika Goyal; Eunice L. Kwak; Janet E. Murphy; David T. Ting; Jennifer Y. Wo; Andrew X. Zhu; Andrew L. Warshaw; Keith D. Lillemoe; Carlos Fernandez-del Castillo

PURPOSE On the basis of the ACCORD trial, FOLFIRINOX is effective in metastatic pancreatic adenocarcinoma (PDAC), making it a rational choice for locally advanced PDAC (LA). Aims of this study are to evaluate the accuracy of imaging in determining the resectability of PDAC and to determine the surgical and clinicopathologic outcomes of pancreatic resections after neoadjuvant FOLFIRINOX therapy. PATIENTS AND METHODS Clinicopathologic data were retrospectively collected for surgical PDAC patients receiving neoadjuvant FOLFIRINOX or no neoadjuvant therapy between April 2011 and February 2014. Americas Hepato-Pancreato-Biliary Association/Society of Surgical Oncology/Society for Surgery of the Alimentary Tract consensus guidelines defined LA and borderline. Imaging was reviewed by a blinded senior pancreatic surgeon. RESULTS Of 188 patients undergoing resection for PDAC, 40 LA/borderline received FOLFIRINOX and 87 received no neoadjuvant therapy. FOLFIRINOX resulted in a significant decrease in tumor size, yet 19 patients were still classified as LA and 9 as borderline. Despite post-FOLFIRINOX imaging suggesting continued unresectability, 92% had an R0 resection. When compared with no neoadjuvant therapy, FOLFIRINOX resulted in significantly longer operative times (393 vs 300 minutes) and blood loss (600 vs 400 mL), but significantly lower operative morbidity (36% vs 63%) and no postoperative pancreatic fistulas. Length of stay (6 vs 7 days), readmissions (20% vs 30%), and mortality were equivalent (1% vs 0%). On final pathology, the FOLFIRINOX group had a significant decrease in lymph node positivity (35% vs 79%) and perineural invasion (72% vs 95%). Median follow-up was 11 months with a significant increase in overall survival with FOLFIRINOX. CONCLUSIONS After neoadjuvant FOLFIRINOX imaging no longer predicts unresectability. Traditional pathologic predictors of survival are improved, and morbidity is decreased in comparison to patients with clearly resectable cancers at the time of presentation.


Clinical Cancer Research | 2013

Targeting the HGF/c-MET Pathway in Hepatocellular Carcinoma

Lipika Goyal; Mandar D. Muzumdar; Andrew X. Zhu

Hepatocellular carcinoma (HCC) is a significant cause of cancer-related morbidity and mortality worldwide. Despite improvements in local therapies, including surgical resection, liver transplantation, and transarterial embolization, the prognosis remains poor for the majority of patients who develop recurrence or present with advanced disease. Systemic therapy with the tyrosine kinase inhibitor sorafenib represents a milestone in advanced HCC but provides a limited survival benefit. Ongoing efforts to study hepatocarcinogenesis have identified an important role for c-MET signaling in the promotion of tumor growth, angiogenesis, and metastasis. In this review, we summarize the preclinical data from human tissue, cell lines, and animal models that implicate c-MET in the pathogenesis of HCC. We also evaluate potential biomarkers that may estimate prognosis or predict response to c-MET inhibitors for more rational clinical trial design. Finally, we discuss the latest clinical trials of c-MET inhibitors in advanced HCC. Clin Cancer Res; 19(9); 2310–8. ©2013 AACR.


Journal of Clinical Oncology | 2016

Multi-Institutional Phase II Study of High-Dose Hypofractionated Proton Beam Therapy in Patients With Localized, Unresectable Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma

Theodore S. Hong; Jennifer Y. Wo; Beow Y. Yeap; Edgar Ben-Josef; Erin McDonnell; Lawrence S. Blaszkowsky; Eunice L. Kwak; Jill N. Allen; Jeffrey W. Clark; Lipika Goyal; Janet E. Murphy; Milind Javle; J Wolfgang; Lorraine C. Drapek; Ronald S. Arellano; Harvey J. Mamon; John T. Mullen; Sam S. Yoon; Kenneth K. Tanabe; Cristina R. Ferrone; David P. Ryan; Thomas F. DeLaney; Christopher H. Crane; Andrew X. Zhu

PURPOSE To evaluate the efficacy and safety of high-dose, hypofractionated proton beam therapy for hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). MATERIALS AND METHODS In this single-arm, phase II, multi-institutional study, 92 patients with biopsy-confirmed HCC or ICC, determined to be unresectable by multidisciplinary review, with a Child-Turcotte-Pugh score (CTP) of A or B, ECOG performance status of 0 to 2, no extrahepatic disease, and no prior radiation received 15 fractions of proton therapy to a maximum total dose of 67.5 Gy equivalent. Sample size was calculated to demonstrate > 80% local control (LC) defined by Response Evaluation Criteria in Solid Tumors (RECIST) 1.0 criteria at 2 years for HCC patients, with the parallel goal of obtaining acceptable precision for estimating outcomes for ICC. RESULTS Eighty-three patients were evaluable: 44 with HCC, 37 with ICC, and two with mixed HCC/ICC. The CTP score was A for 79.5% of patients and B for 15.7%; 4.8% of patients had no cirrhosis. Prior treatment had been given to 31.8% of HCC patients and 61.5% of ICC patients. The median maximum dimension was 5.0 cm (range, 1.9 to 12.0 cm) for HCC patients and 6.0 cm (range, 2.2 to 10.9 cm) for ICC patients. Multiple tumors were present in 27.3% of HCC patients and in 12.8% of ICC patients. Tumor vascular thrombosis was present in 29.5% of HCC patients and in 28.2% of ICC patients. The median dose delivered to both HCC and ICC patients was 58.0 Gy. With a median follow-up among survivors of 19.5 months, the LC rate at 2 years was 94.8% for HCC and 94.1% for ICC. The overall survival rate at 2 years was 63.2% for HCC and 46.5% ICC. CONCLUSION High-dose hypofractionated proton therapy demonstrated high LC rates for HCC and ICC safely, supporting ongoing phase III trials of radiation in HCC and ICC.


Hepatology | 2014

Immune checkpoint blockade in hepatocellular carcinoma: Current progress and future directions

Tai Hato; Lipika Goyal; Tim F. Greten; Dan G. Duda; Andrew X. Zhu

Immune checkpoint blockade has recently emerged as a promising therapeutic approach for various malignancies including hepatocellular carcinoma (HCC). Preclinical and clinical studies have shown the potential benefit of modulating the immunogenicity of HCC. In addition, recent advances in tumor immunology have broadened our understanding of the complex mechanism of immune evasion. In this review we summarize the current knowledge on HCC immunology and discuss the potential of immune checkpoint blockade as a novel HCC therapy from the basic, translational, and clinical perspectives. (Hepatology 2014;60:1776–1782)


Cancer | 2015

Clinical risk factors for the development of hypertension in patients treated with inhibitors of the VEGF signaling pathway

Ole-Petter R. Hamnvik; Toni K. Choueiri; Alexander Turchin; Rana R. McKay; Lipika Goyal; Michael A. Davis; Marina D. Kaymakcalan; Jonathan S. Williams

VEGF signaling pathway inhibitor (anti‐VEGF) therapy is associated with hypertension, but little is known about predisposing clinical characteristics. This study describes the real‐world association between baseline clinical characteristics, blood pressure (BP) response, and survival in patients prescribed anti‐VEGF therapies.


Cancer Discovery | 2017

Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma

Lipika Goyal; Supriya K. Saha; Leah Y. Liu; Giulia Siravegna; Ignaty Leshchiner; Leanne G. Ahronian; Jochen K. Lennerz; Phuong Vu; Vikram Deshpande; Avinash Kambadakone; Benedetta Mussolin; Stephanie Reyes; Laura Henderson; Jiaoyuan Elisabeth Sun; Emily E. Van Seventer; Joseph M. Gurski; Sabrina Baltschukat; Barbara Schacher-Engstler; Louise Barys; Christelle Stamm; Pascal Furet; David P. Ryan; James R. Stone; A. John Iafrate; Gad Getz; Diana Graus Porta; Ralph Tiedt; Alberto Bardelli; Dejan Juric; Ryan B. Corcoran

Genetic alterations in the fibroblast growth factor receptor (FGFR) pathway are promising therapeutic targets in many cancers, including intrahepatic cholangiocarcinoma (ICC). The FGFR inhibitor BGJ398 displayed encouraging efficacy in patients with FGFR2 fusion-positive ICC in a phase II trial, but the durability of response was limited in some patients. Here, we report the molecular basis for acquired resistance to BGJ398 in three patients via integrative genomic characterization of cell-free circulating tumor DNA (cfDNA), primary tumors, and metastases. Serial analysis of cfDNA demonstrated multiple recurrent point mutations in the FGFR2 kinase domain at progression. Accordingly, biopsy of post-progression lesions and rapid autopsy revealed marked inter- and intralesional heterogeneity, with different FGFR2 mutations in individual resistant clones. Molecular modeling and in vitro studies indicated that each mutation led to BGJ398 resistance and was surmountable by structurally distinct FGFR inhibitors. Thus, polyclonal secondary FGFR2 mutations represent an important clinical resistance mechanism that may guide the development of future therapeutic strategies.Significance: We report the first genetic mechanisms of clinical acquired resistance to FGFR inhibition in patients with FGFR2 fusion-positive ICC. Our findings can inform future strategies for detecting resistance mechanisms and inducing more durable remissions in ICC and in the wide variety of cancers where the FGFR pathway is being explored as a therapeutic target. Cancer Discov; 7(3); 252-63. ©2016 AACR.See related commentary by Smyth et al., p. 248This article is highlighted in the In This Issue feature, p. 235.


Clinical Cancer Research | 2016

PD-L1 and HLA Class I Antigen Expression and Clinical Course of the Disease in Intrahepatic Cholangiocarcinoma

Francesco Sabbatino; Vincenzo Villani; Jennifer H. Yearley; Vikram Deshpande; Lei Cai; Ioannis T. Konstantinidis; Christina Moon; Sjoerd P.F.T. Nota; Yangyang Wang; Ahmad Al-Sukaini; Andrew X. Zhu; Lipika Goyal; David T. Ting; Nabeel Bardeesy; Theodore S. Hong; Carlos Fernandez-del Castillo; Kenneth K. Tanabe; Keith D. Lillemoe; Soldano Ferrone; Cristina R. Ferrone

Purpose: More effective therapy is needed for intrahepatic cholangiocarcinoma (ICC). The encouraging clinical results obtained with checkpoint molecule-specific monoclonal antibodies (mAb) have prompted us to investigate whether this type of immunotherapy may be applicable to ICC. The aims of this study were to determine whether (i) patients mount a T-cell immune response to their ICC, (ii) checkpoint molecules are expressed on both T cells and tumor cells, and (iii) tumor cells are susceptible to recognition by cognate T cells. Experimental Design: Twenty-seven ICC tumors were analyzed for (i) lymphocyte infiltrate, (ii) HLA class I and HLA class II expression, and (iii) PD-1 and PD-L1 expression by T cells and ICC cells, respectively. The results of this analysis were correlated with the clinicopathologic characteristics of the patients investigated. Results: Lymphocyte infiltrates were identified in all tumors. PD-L1 expression and HLA class I antigen expression by ICC cells was observed in 8 and 11, respectively, of the 27 tumors analyzed. HLA class I antigen expression correlated with CD8+ T-cell infiltrate. Furthermore, positive HLA class I antigen expression in combination with negative/rare PD-L1 expression was associated with favorable clinical course of the disease. Conclusions: ICC patients are likely to mount a T-cell immune response against their own tumors. Defects in HLA class I antigen expression in combination with PD-L1 expression by ICC cells provide them with an immune escape mechanism. This mechanism justifies the implementation of immunotherapy with checkpoint molecule-specific mAbs in patients bearing ICC tumors without defects in HLA class I antigen expression. Clin Cancer Res; 22(2); 470–8. ©2015 AACR.


Clinical Cancer Research | 2014

Circulating oncometabolite 2-hydroxyglutarate is a potential surrogate biomarker in patients with isocitrate dehydrogenase-mutant intrahepatic cholangiocarcinoma

Darrell R. Borger; Lipika Goyal; Thomas Yau; Ronnie Tung-Ping Poon; Marek Ancukiewicz; Vikram Deshpande; David C. Christiani; Hannah M. Liebman; Hua Yang; Hyeryun Kim; Katharine Yen; Jason E. Faris; A. John Iafrate; Eunice L. Kwak; Jeffrey W. Clark; Jill N. Allen; Lawrence S. Blaszkowsky; Janet E. Murphy; Supriya K. Saha; Theodore S. Hong; Jennifer Y. Wo; Cristina R. Ferrone; Kenneth K. Tanabe; Nabeel Bardeesy; Kimberly Straley; Sam Agresta; David P. Schenkein; Leif W. Ellisen; David P. Ryan; Andrew X. Zhu

Purpose: Mutations in the IDH1 and IDH2 (IDH1/2) genes occur in approximately 20% of intrahepatic cholangiocarcinoma and lead to accumulation of 2-hydroxyglutarate (2HG) in the tumor tissue. However, it remains unknown whether IDH1/2 mutations can lead to high levels of 2HG circulating in the blood and whether serum 2HG can be used as a biomarker for IDH1/2 mutational status and tumor burden in intrahepatic cholangiocarcinoma. Experimental Design: We initially measured serum 2HG concentration in blood samples collected from 31 patients with intrahepatic cholangiocarcinoma in a screening cohort. Findings were validated across 38 resected patients with intrahepatic cholangiocarcinoma from a second cohort with tumor volume measures. Circulating levels of 2HG were evaluated relative to IDH1/2 mutational status, tumor burden, and a number of clinical variables. Results: Circulating levels of 2HG in the screening cohort were significantly elevated in patients with IDH1/2-mutant (median, 478 ng/mL) versus IDH1/2–wild-type (median, 118 ng/mL) tumors (P < 0.001). This significance was maintained in the validation cohort (343 ng/mL vs. 55 ng/mL, P < 0.0001) and levels of 2HG directly correlated with tumor burden in IDH1/2-mutant cases (P < 0.05). Serum 2HG levels ≥170 ng/mL could predict the presence of an IDH1/2 mutation with a sensitivity of 83% and a specificity of 90%. No differences were noted between the allelic variants IDH1 or IDH2 in regard to the levels of circulating 2HG. Conclusions: This study indicates that circulating 2HG may be a surrogate biomarker of IDH1 or IDH2 mutation status in intrahepatic cholangiocarcinoma and that circulating 2HG levels may correlate directly with tumor burden. Clin Cancer Res; 20(7); 1884–90. ©2014 AACR.


Proceedings of the National Academy of Sciences of the United States of America | 2017

An RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinoma

Mark Kalinich; Irun Bhan; Tanya T. Kwan; David T. Miyamoto; Sarah Javaid; Joseph A. LiCausi; John D. Milner; Xin Hong; Lipika Goyal; Srinjoy Sil; Melissa Choz; Uyen Ho; Ravi Kapur; Alona Muzikansky; Huidan Zhang; David A. Weitz; Lecia V. Sequist; David P. Ryan; Raymond T. Chung; Andrew X. Zhu; Kurt J. Isselbacher; David T. Ting; Mehmet Toner; Shyamala Maheswaran; Daniel A. Haber

Significance The early detection of hepatocellular carcinoma (HCC) is of paramount importance for improving patient outcomes, yet an accurate, high-throughput screening methodology has yet to be developed. By combining microfluidic depletion of hematopoietic cells from blood specimens with absolute quantification of lineage-derived transcripts, we demonstrate the highly specific detection of circulating tumor cells, enabling noninvasive detection and clinical monitoring of HCC. Circulating tumor cells (CTCs) are shed into the bloodstream by invasive cancers, but the difficulty inherent in identifying these rare cells by microscopy has precluded their routine use in monitoring or screening for cancer. We recently described a high-throughput microfluidic CTC-iChip, which efficiently depletes hematopoietic cells from blood specimens and enriches for CTCs with well-preserved RNA. Application of RNA-based digital PCR to detect CTC-derived signatures may thus enable highly accurate tissue lineage-based cancer detection in blood specimens. As proof of principle, we examined hepatocellular carcinoma (HCC), a cancer that is derived from liver cells bearing a unique gene expression profile. After identifying a digital signature of 10 liver-specific transcripts, we used a cross-validated logistic regression model to identify the presence of HCC-derived CTCs in nine of 16 (56%) untreated patients with HCC versus one of 31 (3%) patients with nonmalignant liver disease at risk for developing HCC (P < 0.0001). Positive CTC scores declined in treated patients: Nine of 32 (28%) patients receiving therapy and only one of 15 (7%) patients who had undergone curative-intent ablation, surgery, or liver transplantation were positive. RNA-based digital CTC scoring was not correlated with the standard HCC serum protein marker alpha fetoprotein (P = 0.57). Modeling the sequential use of these two orthogonal markers for liver cancer screening in patients with high-risk cirrhosis generates positive and negative predictive values of 80% and 86%, respectively. Thus, digital RNA quantitation constitutes a sensitive and specific CTC readout, enabling high-throughput clinical applications, such as noninvasive screening for HCC in populations where viral hepatitis and cirrhosis are prevalent.


British Journal of Cancer | 2014

Phase II study of gemcitabine, oxaliplatin in combination with panitumumab in KRAS wild-type unresectable or metastatic biliary tract and gallbladder cancer

M S Noel; Jill N. Allen; Thomas Adam Abrams; Matthew B. Yurgelun; Jason E. Faris; Lipika Goyal; Jeffrey W. Clark; Lawrence S. Blaszkowsky; Janet E. Murphy; Hongwu Zheng; Alok A. Khorana; Gregory C. Connolly; Ollivier Hyrien; A Baran; M Herr; Kimmie Ng; Susan Sheehan; D J Harris; Eileen Regan; Darrell R. Borger; Anthony John Iafrate; Charles S. Fuchs; David P. Ryan; Andrew X. Zhu

Background:Current data suggest that platinum-based combination therapy is the standard first-line treatment for biliary tract cancer. EGFR inhibition has proven beneficial across a number of gastrointestinal malignancies; and has shown specific advantages among KRAS wild-type genetic subtypes of colon cancer. We report the combination of panitumumab with gemcitabine (GEM) and oxaliplatin (OX) as first-line therapy for KRAS wild-type biliary tract cancer.Methods:Patients with histologically confirmed, previously untreated, unresectable or metastatic KRAS wild-type biliary tract or gallbladder adenocarcinoma with ECOG performance status 0–2 were treated with panitumumab 6 mg kg−1, GEM 1000 mg m−2 (10 mg m−2 min−1) and OX 85 mg m−2 on days 1 and 15 of each 28-day cycle. The primary objective was to determine the objective response rate by RECIST criteria v.1.1. Secondary objectives were to evaluate toxicity, progression-free survival (PFS), and overall survival.Results:Thirty-one patients received at least one cycle of treatment across three institutions, 28 had measurable disease. Response rate was 45% and disease control rate was 90%. Median PFS was 10.6 months (95% CI 5–24 months) and median overall survival 20.3 months (95% CI 9–25 months). The most common grade 3/4 adverse events were anaemia 26%, leukopenia 23%, fatigue 23%, neuropathy 16% and rash 10%.Conclusions:The combination of gemcitabine, oxaliplatin and panitumumab in KRAS wild type metastatic biliary tract cancer showed encouraging efficacy, additional efforts of genetic stratification and targeted therapy is warranted in biliary tract cancer.

Collaboration


Dive into the Lipika Goyal's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge