Network


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

Hotspot


Dive into the research topics where Melissa Pulitzer is active.

Publication


Featured researches published by Melissa Pulitzer.


The American Journal of Surgical Pathology | 2009

Merkel Cell Polyomavirus Expression in Merkel Cell Carcinomas and Its Absence in Combined Tumors and Pulmonary Neuroendocrine Carcinomas

Achim A. Jungbluth; Natasha Rekthman; Daniel G. Coit; Melissa Pulitzer; Jason Bini; Reety Arora; Nicole Hanson; Jodie Tassello; Denise Frosina; Patrick S. Moore; Yuan Chang

Merkel cell carcinoma (MCC) is the eponym for primary cutaneous neuroendocrine carcinoma. Recently, a new polyoma virus has been identified that is clonally integrated in the genome of the majority of MCCs, with truncating mutations in the viral large T antigen gene. We examined the presence of Merkel cell polyomavirus (MCV) in a set of 17 frozen tumor samples by quantitative polymerase chain reaction; 15 of them (88%) were positive. Sections from corresponding archival material were analyzed by immunohistochemistry (IHC) with the novel monoclonal antibody CM2B4, generated against a predicted antigenic epitope on the MCV T antigen, and tested for the expression of cytokeratin 20 (CK20). Sufficient archival material for IHC was available in only 15 of the 17 cases whose frozen tissue samples had been studied by polymerase chain reaction. Of the 15 tumors analyzed immunohistochemically, 10 (67%) showed positive labeling with CM2B4, 14 (93%) expressed CK20. A tissue microarray of 36 MCCs, 7 combined squamous and neuroendocrine carcinomas of the skin, and 26 pulmonary neuroendocrine carcinomas were also examined by IHC. Of the 36 MCCs assembled on a microarray, 32 (89%) tumors expressed CK20, and 27 (75%) were immunoreactive with CM2B4. The skin tumors with a combined squamous and neuroendocrine phenotype and all pulmonary neuroendocrine carcinomas failed to react with CM2B4. Our study shows that CM2B4 is a useful reagent for the diagnosis of MCC. It labels the majority of MCCs, but fails to react with pulmonary neuroendocrine carcinomas. We also found that neuroendocrine carcinomas of the skin arising in association with a squamous cell carcinoma seem to be independent of MCV.


The American Journal of Surgical Pathology | 2009

Fluorescence in situ hybridization for distinguishing nevoid melanomas from mitotically active nevi

Pedram Gerami; Amanda Wass; Mariam Mafee; Yuaquin Fang; Melissa Pulitzer

Nevoid melanoma may resemble benign compound or intradermal nevi by their silhouette and profile on low power. Higher power usually reveals nuclear atypia, confluence of cells, incomplete maturation and dermal mitotic activity. However, to some extent all of these features maybe seen in benign compound or intradermal nevi and no single criteria can be used to differentiate nevoid melanoma from a benign nevus. The distinction can be particularly problematic in nevi that show mitotic activity and we have noted a recent trend in diagnosis of melanocytic neoplasms with dermal mitosis as nevoid melanoma despite the presence of normal maturation in the dermis and lack of significant nuclear atypia. Therefore in this study we evaluated 10 nevoid melanomas, 4 of which resulted in metastasis and 10 mitotically active nevi with fluorescence in situ hybridization targeting key chromosomal loci previously shown to effectively discriminate been malignant and benign melanocytic neoplasms. All 10 nevoid melanomas showed copy number abnormalities by fluorescence in situ hybridization in either chromosome 6 or 11 while none of the 10 mitotically active nevi did. The results demonstrate that fluorescence in situ hybridization targeting key chromosomal loci on chromosomes 6 and 11 can be effective in discriminating nevoid melanomas from mitotically active nevi. Additionally, our study presents further evidence that dermal mitoses alone without other diagnostic features such as nuclear atypia and lack of maturation does not constitute sufficient evidence alone for a diagnosis of melanoma.


Annals of Surgery | 2011

Five hundred patients with Merkel cell carcinoma evaluated at a single institution.

Ryan C. Fields; Joanne F. Chou; Katherine S. Panageas; Melissa Pulitzer; Peter J. Allen; Dennis H. Kraus; Mary S. Brady; Daniel G. Coit

Objective:To identify factors associated with survival in Merkel cell carcinoma (MCC). Background:Merkel cell carcinoma is a rare cutaneous neoplasm. Staging and treatment are based on studies, which incompletely characterize the disease. Methods:Review of a prospective database was performed. Overall survival (OS) was estimated by the Kaplan-Meier method. Disease-specific death (DSD) was analyzed by the competing risks method. Factors associated with OS and DSD were determined by the log-rank test and Grays test, respectively. Results:A total of 500 patients with MCC treated at our institution from 1969 to 2010 were identified. Eighty-eight patients presented older than 6 months after diagnosis and were excluded from further analysis. Of the remaining 412 patients, the median age at diagnosis was 71 years. Median follow-up was 3 years. Fifty percent of patients died during follow-up: 25% died of disease, 24% died of other causes. Five-year OS and DSD were 56% and 30%, respectively. Pathologic stage and lymphovascular invasion were independent predictors of DSD. Patients with metastatic disease (stage 4) or clinically positive lymph nodes (stage 3b) had increased DSD compared with patients with microscopically positive (stage 3a) or negative lymph nodes (stage 1 and 2). There was no difference in DSD between stage 3a or 2 compared with stage 1. Importantly, only 1 of 132 patients without lymphovascular invasion died of MCC. Conclusions:OS is a poor measure of the influence of MCC on life expectancy. The presence of lymphovascular invasion and clinically, but not microscopically, positive lymph nodes were associated with increased DSD. These factors should be incorporated into MCC staging and treatment recommendations.


Journal of Clinical Oncology | 2015

Phase II Investigator-Initiated Study of Brentuximab Vedotin in Mycosis Fungoides and Sézary Syndrome With Variable CD30 Expression Level: A Multi-Institution Collaborative Project

Youn H. Kim; Mahkam Tavallaee; Uma Sundram; Katrin A. Salva; Gary S. Wood; Shufeng Li; Sima Rozati; Seema Nagpal; Michael Krathen; Sunil Reddy; Richard T. Hoppe; Annie Nguyen-Lin; Wen-Kai Weng; Randall Armstrong; Melissa Pulitzer; Ranjana H. Advani; Steven M. Horwitz

PURPOSE In contrast to Hodgkin lymphoma and systemic anaplastic large-cell lymphoma, CD30 expression of malignant lymphocytes in mycosis fungoides (MF) and Sézary syndrome (SS) is quite variable. Clinical activity and safety of brentuximab vedotin, a CD30 targeting antibody-drug conjugate, was evaluated in MF and SS. Tissue and blood biomarkers of clinical response were explored. PATIENTS AND METHODS In this phase II study, patients with MF or SS with negligible to 100% CD30 expression levels were treated with brentuximab vedotin (1.8 mg/kg) every 3 weeks for a maximum of sixteen doses. The primary end point was overall global response rate. Secondary end points included correlation of tissue CD30 expression level with clinical response, time to response, duration of response, progression-free and event-free survivals, and safety. RESULTS Of the 32 patients enrolled and treated, 30 patients had available efficacy evaluations. Objective global response was observed in 21 (70%) of 30 patients (90% CI, 53% to 83%). CD30 expression assessed by immunohistochemistry was highly variable, with a median CD30max of 13% (range, 0% to 100%). Those with <5% CD30 expression had a lower likelihood of global response than did those with 5% or greater CD30 expression (P < .005). CD163 positive tumor-associated macrophages, many of which coexpress CD30, were abundant in tissue. Peripheral neuropathy was the most common adverse event. CONCLUSION Brentuximab vedotin demonstrated significant clinical activity in treatment-refractory or advanced MF or SS with a wide range of CD30 expression levels. Additional biomarker studies may help optimize rational design of combination therapies with brentuximab vedotin.


Clinical Cancer Research | 2013

Phase II trial of MEK inhibitor selumetinib (AZD6244, ARRY-142886) in patients with BRAFV600E/K- mutated melanoma

Federica Catalanotti; David B. Solit; Melissa Pulitzer; Michael F. Berger; Sasinya N. Scott; Tunc Iyriboz; Mario E. Lacouture; Katherine S. Panageas; Jedd D. Wolchok; Richard D. Carvajal; Gary K. Schwartz; Neal Rosen; Paul B. Chapman

Purpose: Test the hypothesis that in BRAF-mutated melanomas, clinical responses to selumetinib, a MEK inhibitor, will be restricted to tumors in which the PI3K/AKT pathway is not activated. Experimental Design: We conducted a phase II trial in patients with melanoma whose tumors harbored a BRAF mutation. Patients were stratified by phosphorylated-AKT (pAKT) expression (high vs. low) and treated with selumetinib 75 mg per os twice daily. Pretreatment tumors were also analyzed for genetic changes in 230 genes of interest using an exon-capture approach. Results: The high pAKT cohort was closed after no responses were seen in the first 10 patients. The incidence of low pAKT melanoma tumors was low (∼25% of melanomas tested) and this cohort was eventually closed because of poor accrual. However, among the five patients with melanoma accrued in the low pAKT cohort, there was one partial response (PR). Two other patients had near PRs before undergoing surgical resection of residual disease (one patient) or discontinuation of treatment due to toxicity (one patient). Among the two nonresponding, low pAKT patients with melanoma, co-mutations in MAP2K1, NF1, and/or EGFR were detected. Conclusions: Tumor regression was seen in three of five patients with BRAF-mutated, low pAKT melanomas; no responses were seen in the high pAKT cohort. These results provide rationale for co-targeting MEK and PI3K/AKT in patients with BRAF mutant melanoma whose tumors express high pAKT. However, the complexity of genetic changes in melanoma indicates that additional genetic information will be needed for optimal selection of patients likely to respond to MEK inhibitors. Clin Cancer Res; 19(8); 2257–64. ©2013 AACR.


Advances in Anatomic Pathology | 2009

Merkel cell carcinoma: review.

Melissa Pulitzer; Bijal Amin

Merkel cell carcinoma (MCC) is synonymous with primary cutaneous neuroendocrine carcinoma. It tends to affects elderly whites, but there is also an increased incidence among immunosuppressed patients. The recent identification of a novel polyomavirus associated with the tumor has stimulated renewed interest in its pathogenesis. MCC tends to show classic histologic features of a neuroendocrine carcinoma and is often positive for CK20, but nonclassic cytologic findings and unusual immunophenotypes may be observed and can lead to a diagnostic confusion. MCC needs to be distinguished from other primary cutaneous tumors with a small cell appearance and metastatic tumors. Surgical excision is the treatment of choice, but radiation therapy has also found to be effective. Sentinel lymph node biopsy has become an integral part of the staging of patients with MCC.


The American Journal of Surgical Pathology | 2013

Immunohistochemical analysis of BRAFV600E expression of primary and metastatic melanoma and comparison with mutation status and melanocyte differentiation antigens of metastatic lesions

Cyrus V. Hedvat; Melissa Pulitzer; Andreas von Deimling; Achim A. Jungbluth

BRAFV600E is the most common mutation in cutaneous melanoma and has become the target of treatment for patients with metastatic melanoma. A number of methods are currently available to determine mutation status. Recently, a monoclonal antibody (VE1) against mutant BRAFV600E was generated. Its use permits assessment of the mutant protein expression throughout a tumor sample and may allow faster and cheaper determination of the mutation status in selected cases. However, for BRAFV600E protein expression analysis to be of clinical value, high sensitivity and specificity of the antibody is a prerequisite. In this study we analyzed 44 metastatic melanoma samples with a known BRAFV600E mutation status with immunohistochemical expression of the BRAFV600E protein. None of the 22 tumors that lacked the BRAFV600E mutation labeled with the antibody VE1. This set of VE1-immunonegative tumors included 4 metastatic lesions with the BRAFV600K mutation. All 22 tumor samples that were known to carry the BRAFV600E mutation were immunoreactive with VE1. Sixteen of them stained strongly and homogenously throughout the tumor sample. However, 6 tumor samples contained both BRAFV600E-immunopositive and BRAFV600E-immunonegative cell populations. When the BRAF status was compared with immunoreactivity for melanocyte differentiation antigens, no significant difference in the expression of melan-A, microphthalmia transcription factor, gp100, or tyrosinase was found between mutant and wild-type tumors. In addition to metastatic lesions, we also examined 20 primary melanomas for the expression of BRAFV600E. Seven of 10 superficial spreading melanomas were immunoreactive with the antibody VE1. Five tumors were strongly and homogenously immunoreactive. In 2 primary tumors the staining was focal, involving only a subpopulation of the tumor. None of the nonsuperficial spreading melanomas was immunoreactive. In 7 primary tumors the mutation status could be analyzed: only tumors carrying the BRAFV600E mutation were immunoreactive with VE1. The high specificity and sensitivity of VE1 for the detection of mutant BRAFV600E suggests a valuable reagent for clinical purposes. Heterogeneity in BRAF expression may be relevant for treatment response to BRAF inhibitors.


The American Journal of Surgical Pathology | 2009

Atypical Spitzoid Melanocytic Tumors With Positive Sentinel Lymph Nodes in Children and Teenagers, and Comparison With Histologically Unambiguous and Lethal Melanomas

Rajmohan Murali; Melissa Pulitzer; Stanley W. McCarthy; John F. Thompson; Helen M. Shaw; Mary S. Brady; Daniel G. Coit; Stephen W. Dusza; James S. Wilmott; Marc Kayton; Michael P. LaQuaglia; Richard A. Scolyer

Children and teenagers with a positive sentinel lymph node (SLN) after a prior diagnosis of an atypical spitzoid melanocytic tumor (ASMT) are usually cared for clinically in the same way as patients with melanoma. Little is known about long-term follow-up of these individuals to determine whether this practice is appropriate. To learn more about the biology of these tumors we retrospectively reviewed the clinical and pathologic findings of children and teenagers (<18 y of age at the time of diagnosis) with an ASMT, positive SLN and follow-up of at least 3 years. Their findings were compared with histologically unambiguous melanomas of children or teenagers, who had a positive SLN or died of metastatic melanoma. Eleven individuals, 6 girls and 5 boys, with primary ASMT and positive SLN were identified. The primary tumors ranged in thickness from 2.1 to 12 mm (median, 4.6 mm; mean, 5 mm). The tumor mitotic rate ranged from 1 to 10 mitoses/mm2 (median, 3/mm2, median, 3/mm2). The positive SLNs included 6 nodes with intranodal melanocytic aggregates measuring <1 mm in greatest dimension, and 5 nodes, in which the size of the melanocyte deposits was ≥1 mm. All the patients with ASMT and positive SLN remained free of disease with a median follow-up of 47 months (mean, 61 mo, range: 36 to 132 mo). In contrast, 2 of 5 patients <18 years of age with a histologically unambiguous melanoma and a positive SLN died of metastatic melanoma. The overall disease-specific mortality rate for all patients <18 years of age diagnosed with melanoma was 12%. Our findings confirm that children and teenagers with ASMTs and positive SLNs have a less aggressive clinical course than those with histologically unambiguous melanoma.


Journal of The American Academy of Dermatology | 2013

The risk of rash associated with ipilimumab in patients with cancer: A systematic review of the literature and meta-analysis

Kira Minkis; Benjamin C. Garden; Shenhong Wu; Melissa Pulitzer; Mario E. Lacouture

BACKGROUND Ipilimumab is a human antibody that inhibits cytotoxic T-lymphocyte-associated antigen 4, leading to increases in T-cell activation and interleukin 2 secretion and has been approved for the treatment of advanced melanoma. Dermatologic adverse events such as rash, pruritus, and vitiligo have been reported in trials, with varying incidences. The overall incidence and risk of rash to ipilimumab is unknown. OBJECTIVE We conducted a systematic review of the literature and performed a meta-analysis to ascertain the incidence and risk of developing rash among patients receiving ipilimumab. METHODS Databases from PubMed and Web of Science from January 1998 until July 2011 and abstracts presented at the American Society of Clinical Oncology meetings from 2004 through 2011 were searched to identify relevant studies. The incidence and relative risk of rash were calculated using random effects or fixed effects model depending on the heterogeneity of included studies. RESULTS A total of 1208 patients from clinical trials were included in this analysis. The overall incidence of all-grade rash was 24.3% (95% confidence interval [CI] 21.4%-27.6%), with a relative risk of 4.00 (95% CI 2.63-6.08, P < .001). The overall incidence of high-grade rash was 2.4% (95% CI 1.1%-5.1%), with a relative risk of 3.31 (95% CI 0.70-15.76, P = .13). LIMITATIONS The ability to detect rash may vary among institutions. CONCLUSION There is a significant risk of developing rash in patients with cancer receiving ipilimumab. There was no statistically significant difference in the risk of rash based on dose or underlying tumor. Adequate monitoring and early intervention are recommended to prevent decreased quality of life and inconsistent dosing.


Journal of Cutaneous Pathology | 2010

Distinction of conjunctival melanocytic nevi from melanomas by fluorescence in situ hybridization

Yuqiang Fang; Suresh C. Jhanwar; Melissa Pulitzer; Brian P. Marr; David H. Abramson

A conjunctival melanocytic nevus may on occasion be difficult to distinguish from melanoma both clinically and histopathologically. An unambiguous correct diagnosis is critical because of major differences in management and prognosis. We evaluated a fluorescence in situ hybridization (FISH) assay, which has previously been shown to be of value for the diagnosis of melanocytic nevi and melanomas of the skin, using probes targeting 6p25 (RREB1), 6q23 (MYB), 11q13 (CCND1) and centromere 6 (CEP6), for its potential to assist in the distinction of conjunctival melanocytic nevi from melanomas. Four melanocytic nevi and eight melanomas of the conjunctiva were analyzed. Two of the melanomas were diagnostically problematic because of suboptimal histopathology. None of the conjunctival melanocytic nevi showed a level of chromosomal aberrations that met FISH criteria for a diagnosis of melanoma. All eight conjunctival melanomas (six unequivocal and two suspicious lesions) met FISH criteria for melanoma. Thus, results from FISH assay targeting 6p25, 6q23, 11q13 and centromere 6 correlated well with the histopathologic diagnoses and supported the histopathologic suspicion in two problem cases. The findings encourage further exploration of this technique as an ancillary method for the work‐up of conjunctival melanocytic proliferations.

Collaboration


Dive into the Melissa Pulitzer's collaboration.

Top Co-Authors

Avatar

Patricia L. Myskowski

NewYork–Presbyterian Hospital

View shared research outputs
Top Co-Authors

Avatar

Steven M. Horwitz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Christiane Querfeld

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alison J. Moskowitz

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mario E. Lacouture

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shamir Geller

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Daniel G. Coit

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Michael A. Marchetti

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Paul B. Chapman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge