Network


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

Hotspot


Dive into the research topics where Muzaffar Akram is active.

Publication


Featured researches published by Muzaffar Akram.


Modern Pathology | 2005

Epidermal growth factor receptor expression and gene amplification in colorectal carcinoma: An immunohistochemical and chromogenic in situ hybridization study

Jinru Shia; David S. Klimstra; Allan R. Li; Jing Qin; Leonard Saltz; Julie Teruya-Feldstein; Muzaffar Akram; Ki Young Chung; Davis Yao; Philip B. Paty; William L. Gerald; Beiyun Chen

Recent data suggest that detection of epidermal growth factor receptor protein by immunohistochemistry (IHC) does not predict response to the antiepidermal growth factor receptor drug, cetuximab, in patients with colorectal carcinoma. In searching for foundation for further investigation to optimize patient selection for cetuximab therapy, this study sought to exploit the tissue microarray and chromogenic in situ hybridization techniques to evaluate the status of epidermal growth factor receptor gene amplification in colorectal cancer and its relationship with protein expression by IHC. The study included 158 primary or metastatic colorectal adenocarcinomas. Immunohistochemical results were scored as 0–3+ based on the intensity of membrane staining. The in situ hybridization signals were counted in 30 nuclei per tissue core. Overall, the rate of tissue loss was 7%, yielding 147 analyzable cases: 123 primary, 24 metastatic. Positive immunohistochemical staining of any intensity was detected in 85% (105/123) of primary and 79% (19/24) of metastatic tumors, whereas gene amplification (>5 gene copies/nucleus) was only seen in 12% (15/123) of primary and 8% (2/24) of metastatic tumors. Only 2/15 primary and 1/2 metastatic tumors that showed gene amplification were amplified at a high level (>10 gene copies/nucleus). Although a positive correlation was detected between the intensity of protein expression and the likelihood of gene amplification in both the primary (P=0.01) and the metastatic (P=0.05) tumors, IHC had a low specificity (17% in primary, 23% in metastatic) in predicting gene amplification. Conversely, all tumors that did not express the protein by IHC lacked gene amplification. Thus, this study shows that only a small fraction of epidermal growth factor receptor- positive colorectal carcinomas detected by IHC are associated with gene amplification. Additional studies are needed to determine whether epidermal growth factor receptor gene amplification bears any informative value in predicting response to cetuximab-based therapy.


The American Journal of Surgical Pathology | 2005

Expression of WT1, CA 125, and GCDFP-15 as useful markers in the differential diagnosis of primary ovarian carcinomas versus metastatic breast cancer to the ovary.

Carmen Tornos; Robert A. Soslow; Shirley Chen; Muzaffar Akram; Amanda J. Hummer; Nadeen Abu-Rustum; Larry Norton; Lee K. Tan

Metastatic breast carcinoma to the ovary is sometimes difficult to differentiate from primary ovarian carcinoma. This problem is often encountered in breast carcinoma patients who develop adnexal masses. ER and PR can be positive in a high percentage of breast and ovarian carcinomas, and therefore cannot be used in the differential diagnosis of these entities. WT1 and CA125 have been identified as possible markers for ovarian cancer. However, no studies have been done that specifically compare the immunophenotype of breast carcinoma metastatic to ovary with that of primary ovarian cancer. Thirty-nine cases of metastatic breast carcinoma to the ovary, 36 primary breast carcinomas, and 42 primary ovarian carcinomas were examined immunohistochemically for the expression of WT1, CA125, carcinoembryonic antigen, MUC2, MUC1, and GCDFP. The percentage of cells stained and the intensity of staining were recorded. Thirty-two ovarian carcinomas (76%) were positive for WT1, including 31 of 33 (94%) serous carcinomas. Most of them had strong and diffuse staining. None of the breast cancers either primary or metastatic to the ovary expressed WT1. Thirty-eight (90%) ovarian carcinomas were positive for CA125, most of them with strong and diffuse staining. Most breast carcinomas were negative for CA125, with only 6 (16%) of the primary ones and 5 (12%) of the metastatic showing weak and focal positivity. All ovarian carcinomas were negative for GCDFP. Five primary breast cancers (14%) and 17 (43%) metastatic to the ovary were positive for GCDFP. Nine (21%) ovarian carcinomas, 8 (22%) primary breast carcinomas, and 13 (33%) metastatic to the ovary were positive for carcinoembryonic antigen. Almost all tumors examined were positive for MUC1 (100% ovarian carcinomas, 100% primary breast carcinomas, and 95% metastatic breast carcinomas to ovary). MUC2 was positive in 10 (24%) ovarian carcinomas, 3 (8%) primary breast cancers, and 12 (30%) metastases to the ovary. The presence of immunoreactivity for WT1 and CA125 in a carcinoma involving ovary strongly favors a primary lesion. Most ovarian carcinomas are positive for both markers, whereas the majority of metastatic breast carcinomas to the ovary are negative. GCDFP can be complementary in this differential diagnosis.


Cancer | 2013

Classic lobular carcinoma in situ and atypical lobular hyperplasia at percutaneous breast core biopsy: outcomes of prospective excision.

Melissa P. Murray; Chad Luedtke; Laura Liberman; Tatjana Nehhozina; Muzaffar Akram; Edi Brogi

No consensus exists on the need to excise breast lesions that yield classic lobular carcinoma in situ (LCIS) or atypical lobular hyperplasia (ALH) (known together as classic lobular neoplasia [LN]) as the highest risk lesion at percutaneous core‐needle biopsy (CNB). Here, the authors report findings from 72 consecutive lesions with LN at CNB and prospective surgical excision (EXB).


The American Journal of Surgical Pathology | 2011

Intracystic papillary carcinoma of the breast: An in situ or invasive tumor? Results of immunohistochemical analysis and clinical follow-up.

Christine A. Wynveen; Tatjana Nehhozina; Muzaffar Akram; Mohammed Hassan; Larry Norton; Kimberly J. Van Zee; Edi Brogi

Intracystic papillary carcinoma (IPC) is regarded as an intraductal neoplasm, but recent evidence suggests that it could be invasive, as it often lacks myoepithelial lining. We evaluated myoepithelial cells and collagen IV, a basement membrane component, in 40 IPCs from 39 (35 female and 4 male) patients and assessed their clinical management and follow-up. The mean patient age at diagnosis was 68 years, and the mean tumor size was 1.8 cm. Thirteen cases were pure IPC, 8 cases were IPC with or without microinvasion, and 19 cases were IPC with invasive carcinoma (IPC+IC), including 1 mucinous and 1 cribriform carcinoma. Ductal carcinoma in situ associated more often with IPC+IC (84.2%) than with pure IPC (61.5%) or IPC with or without microinvasion (62.5%). Myoepithelial cells were completely absent in 33 of 40 (82.5%) IPCs, and only focal in the remaining 7 of 40 cases (17.5%). Collagen IV lining was discontinuous in most cases (89%). All tumors were estrogen receptor positive and HER2 negative; most were progesterone receptor positive (93%). Eleven patients underwent mastectomy and 28 lumpectomy; 3 of 27 (11%) patients had lymph node involvement. Fourteen of all patients treated with breast conservation received radiation, 10 hormonal treatment, and none chemotherapy. Four patients treated conservatively (3 with pure IPC and 1 with IPC+IC) recurred locally, including one who later developed bone metastasis. We conclude that IPC constitutes a spectrum of intraductal and IC, with predominance of the latter. IPC rarely involves lymph nodes and carries very good prognosis, but can recur locally. This type of tumor is strongly estrogen receptor positive and hormonal therapy should be pursued for its management, whereas the benefit of radiation after lumpectomy remains unclear.


Clinical Cancer Research | 2016

The Genomic Landscape of Male Breast Cancers

Salvatore Piscuoglio; Charlotte K.Y. Ng; Melissa P. Murray; Elena Guerini-Rocco; Luciano G. Martelotto; Felipe C. Geyer; François-Clément Bidard; Samuel H. Berman; Nicola Fusco; Rita A. Sakr; Carey A. Eberle; Leticia De Mattos-Arruda; Gabriel S. Macedo; Muzaffar Akram; Timour Baslan; James Hicks; Tari A. King; Edi Brogi; Larry Norton; Britta Weigelt; Clifford A. Hudis; Jorge S. Reis-Filho

Purpose: Male breast cancer is rare, and its genomic landscape has yet to be fully characterized. Lacking studies in men, treatment of males with breast cancer is extrapolated from results in females with breast cancer. We sought to define whether male breast cancers harbor somatic genetic alterations in genes frequently altered in female breast cancers. Experimental Design: All male breast cancers were estrogen receptor–positive, and all but two were HER2-negative. Fifty-nine male breast cancers were subtyped by immunohistochemistry, and tumor–normal pairs were microdissected and subjected to massively parallel sequencing targeting all exons of 241 genes frequently mutated in female breast cancers or DNA-repair related. The repertoires of somatic mutations and copy number alterations of male breast cancers were compared with that of subtype-matched female breast cancers. Results: Twenty-nine percent and 71% of male breast cancers were immunohistochemically classified as luminal A–like or luminal B–like, respectively. Male breast cancers displayed a heterogeneous repertoire of somatic genetic alterations that to some extent recapitulated that of estrogen receptor (ER)-positive/HER2-negative female breast cancers, including recurrent mutations affecting PIK3CA (20%) and GATA3 (15%). ER-positive/HER2-negative male breast cancers, however, less frequently harbored 16q losses, and PIK3CA and TP53 mutations than ER-positive/HER2-negative female breast cancers. In addition, male breast cancers were found to be significantly enriched for mutations affecting DNA repair–related genes. Conclusions: Male breast cancers less frequently harbor somatic genetic alterations typical of ER-positive/HER2-negative female breast cancers, such as PIK3CA and TP53 mutations and losses of 16q, suggesting that at least a subset of male breast cancers are driven by a distinct repertoire of somatic changes. Given the genomic differences, caution may be needed in the application of biologic and therapeutic findings from studies of female breast cancers to male breast cancers. Clin Cancer Res; 22(16); 4045–56. ©2016 AACR.


Cell | 2016

Multi-organ Site Metastatic Reactivation Mediated by Non-canonical Discoidin Domain Receptor 1 Signaling

Hua Gao; Goutam Chakraborty; Zhanguo Zhang; Intissar Akalay; Mayur Gadiya; Yaquan Gao; Surajit Sinha; Jian Hu; Cizhong Jiang; Muzaffar Akram; Edi Brogi; Birgit Leitinger; Filippo G. Giancotti

Genetic screening identifies the atypical tetraspanin TM4SF1 as a strong mediator of metastatic reactivation of breast cancer. Intriguingly, TM4SF1 couples the collagen receptor tyrosine kinase DDR1 to the cortical adaptor syntenin 2 and, hence, to PKCα. The latter kinase phosphorylates and activates JAK2, leading to the activation of STAT3. This non-canonical mechanism of signaling induces the expression of SOX2 and NANOG; sustains the manifestation of cancer stem cell traits; and drives metastatic reactivation in the lung, bone, and brain. Bioinformatic analyses and pathological studies corroborate the clinical relevance of these findings. We conclude that non-canonical DDR1 signaling enables breast cancer cells to exploit the ubiquitous interstitial matrix component collagen I to undergo metastatic reactivation in multiple target organs.


The Journal of Pathology | 2016

Massively parallel sequencing of phyllodes tumours of the breast reveals actionable mutations, and TERT promoter hotspot mutations and TERT gene amplification as likely drivers of progression

Salvatore Piscuoglio; Charlotte K.Y. Ng; Melissa P. Murray; Kathleen A. Burke; Marcia Edelweiss; Felipe C. Geyer; Gabriel S. Macedo; Akiko Inagaki; Anastasios D. Papanastasiou; Luciano G. Martelotto; Caterina Marchiò; Raymond S. Lim; Rafael A. Ioris; Pooja K. Nahar; Ino de Bruijn; Lillian Mary Smyth; Muzaffar Akram; Dara S. Ross; John H.J. Petrini; Larry Norton; David B. Solit; José Baselga; Edi Brogi; Marc Ladanyi; Britta Weigelt; Jorge S. Reis-Filho

Phyllodes tumours (PTs) are breast fibroepithelial lesions that are graded based on histological criteria as benign, borderline or malignant. PTs may recur locally. Borderline PTs and malignant PTs may metastasize to distant sites. Breast fibroepithelial lesions, including PTs and fibroadenomas, are characterized by recurrent MED12 exon 2 somatic mutations. We sought to define the repertoire of somatic genetic alterations in PTs and whether these may assist in the differential diagnosis of these lesions. We collected 100 fibroadenomas, 40 benign PTs, 14 borderline PTs and 22 malignant PTs; six, six and 13 benign, borderline and malignant PTs, respectively, and their matched normal tissue, were subjected to targeted massively parallel sequencing (MPS) using the MSK‐IMPACT sequencing assay. Recurrent MED12 mutations were found in 56% of PTs; in addition, mutations affecting cancer genes (eg TP53, RB1, SETD2 and EGFR) were exclusively detected in borderline and malignant PTs. We found a novel recurrent clonal hotspot mutation in the TERT promoter (−124 C>T) in 52% and TERT gene amplification in 4% of PTs. Laser capture microdissection revealed that these mutations were restricted to the mesenchymal component of PTs. Sequencing analysis of the entire cohort revealed that the frequency of TERT alterations increased from benign (18%) to borderline (57%) and to malignant PTs (68%; p < 0.01), and TERT alterations were associated with increased levels of TERT mRNA (p < 0.001). No TERT alterations were observed in fibroadenomas. An analysis of TERT promoter sequencing and gene amplification distinguished PTs from fibroadenomas with a sensitivity and a positive predictive value of 100% (CI 95.38–100%) and 100% (CI 85.86–100%), respectively, and a sensitivity and a negative predictive value of 39% (CI 28.65–51.36%) and 68% (CI 60.21–75.78%), respectively. Our results suggest that TERT alterations may drive the progression of PTs, and may assist in the differential diagnosis between PTs and fibroadenomas. Copyright


Modern Pathology | 2016

Genetic alterations of triple negative breast cancer by targeted next-generation sequencing and correlation with tumor morphology

Paul S Weisman; Charlotte K.Y. Ng; Edi Brogi; Rachel E Eisenberg; Helen H. Won; Salvatore Piscuoglio; Maria Rosaria De Filippo; Rafael A. Ioris; Muzaffar Akram; Larry Norton; Britta Weigelt; Michael F. Berger; Jorge S. Reis-Filho; Hannah Y. Wen

Triple negative breast cancer represents a heterogeneous group of breast carcinomas, both at the histologic and genetic level. Although recent molecular studies have comprehensively characterized the genetic landscape of these tumors, few have integrated a detailed histologic examination into the analysis. In this study, we defined the genetic alterations in 39 triple negative breast cancers using a high-depth targeted massively parallel sequencing assay and correlated the findings with a detailed morphologic analysis. We obtained representative frozen tissue of primary triple negative breast cancers from patients treated at our institution between 2002 and 2010. We characterized tumors according to their histologic subtype and morphologic features. DNA was extracted from paired frozen primary tumor and normal tissue samples and was subjected to a targeted massively parallel sequencing platform comprising 229 cancer-associated genes common across all experiments. The average number of non-synonymous mutations was 3 (range 0–10) per case. The most frequent somatic alterations were mutations in TP53 (74%) and PIK3CA (10%) and MYC amplifications (26%). Triple negative breast cancers with apocrine differentiation less frequently harbored TP53 mutations (25%) and MYC gains (0%), and displayed a high mutation frequency in PIK3CA and other PI3K signaling pathway-related genes (75%). Using a targeted massively parallel sequencing platform, we identified the key somatic genetic alterations previously reported in triple negative breast cancers. Furthermore, our findings show that triple negative breast cancers with apocrine differentiation constitute a distinct subset, characterized by a high frequency of PI3K pathway alterations similar to luminal subtypes of breast cancer.


The Journal of Pathology | 2016

Microglandular adenosis associated with triple-negative breast cancer is a neoplastic lesion of triple-negative phenotype harbouring TP53 somatic mutations

Elena Guerini-Rocco; Salvatore Piscuoglio; Charlotte K.Y. Ng; Felipe C. Geyer; Maria Rosaria De Filippo; Carey A. Eberle; Muzaffar Akram; Nicola Fusco; Shu Ichihara; Rita A. Sakr; Yasushi Yatabe; Anne Vincent-Salomon; Emad A. Rakha; Ian O. Ellis; Y Hannah Wen; Britta Weigelt; Stuart J. Schnitt; Jorge S. Reis-Filho

Microglandular adenosis (MGA) is a rare proliferative lesion of the breast composed of small glands lacking myoepithelial cells and lined by S100‐positive, oestrogen receptor (ER)‐negative, progesterone receptor (PR)‐negative, and HER2‐negative epithelial cells. There is evidence to suggest that MGA may constitute a non‐obligate precursor of triple‐negative breast cancer (TNBC). We sought to define the genomic landscape of pure MGA and of MGA, atypical MGA (AMGA) and associated TNBCs, and to determine whether synchronous MGA, AMGA, and TNBCs would be clonally related. Two pure MGAs and eight cases of MGA and/or AMGA associated with in situ or invasive TNBC were collected, microdissected, and subjected to massively parallel sequencing targeting all coding regions of 236 genes recurrently mutated in breast cancer or related to DNA repair. Pure MGAs lacked clonal non‐synonymous somatic mutations and displayed limited copy number alterations (CNAs); conversely, all MGAs (n = 7) and AMGAs (n = 3) associated with TNBC harboured at least one somatic non‐synonymous mutation (range 3–14 and 1–10, respectively). In all cases where TNBCs were analyzed, identical TP53 mutations and similar patterns of gene CNAs were found in the MGA and/or AMGA and in the associated TNBC. In the MGA/AMGA associated with TNBC lacking TP53 mutations, somatic mutations affecting PI3K pathway‐related genes (eg PTEN, PIK3CA, and INPP4B) and tyrosine kinase receptor signalling‐related genes (eg ERBB3 and FGFR2) were identified. At diagnosis, MGAs associated with TNBC were found to display subclonal populations, and clonal shifts in the progression from MGA to AMGA and/or to TNBC were observed. Our results demonstrate the heterogeneity of MGAs, and that MGAs associated with TNBC, but not necessarily pure MGAs, are genetically advanced, clonal, and neoplastic lesions harbouring recurrent mutations in TP53 and/or other cancer genes, supporting the notion that a subset of MGAs and AMGAs may constitute non‐obligate precursors of TNBCs. Copyright


PLOS ONE | 2014

Mesothelin expression in triple negative breast carcinomas correlates significantly with basal-like phenotype, distant metastases and decreased survival.

Gary Tozbikian; Edi Brogi; Kyuichi Kadota; Jeffrey Catalano; Muzaffar Akram; Sujata Patil; Alice Y. Ho; Jorge S. Reis-Filho; Britta Weigelt; Larry Norton; Prasad S. Adusumilli; Hannah Yong Wen

Mesothelin is a cell surface associated antigen expressed on mesothelial cells and in some malignant neoplasms. Mesothelin-targeted therapies are in phase I/II clinical trials. The clinicopathologic and prognostic significance of mesothelin expression in triple negative breast carcinomas (TNBC) has not been fully assessed. We evaluated the expression of mesothelin and of basal markers in tissue microarrays of 226 TNBC and 88 non-TNBC and assessed the clinicopathologic features of mesothelin-expressing breast carcinomas. Furthermore, we investigated the impact of mesothelin expression on the disease-free and overall survival of patients with TNBC. We found that mesothelin expression is significantly more frequent in TNBC than in non-TNBC (36% vs 16%, respectively; p = 0.0006), and is significantly correlated with immunoreactivity for basal keratins, but not for EGFR. Mesothelin-positive and mesothelin-negative TNBC were not significantly different by patients’ race, tumor size, histologic grade, tumor subtype, lymphovascular invasion and lymph node metastases. Patients with mesothelin-positive TNBC were older than patients with mesothelin-negative TNBC, developed more distant metastases with a shorter interval, and had significantly lower overall and disease-free survival. Based on our results, patients with mesothelin-positive TNBC could benefit from mesothelin-targeted therapies.

Collaboration


Dive into the Muzaffar Akram's collaboration.

Top Co-Authors

Avatar

Edi Brogi

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Larry Norton

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Britta Weigelt

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jorge S. Reis-Filho

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sujata Patil

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Adriana D. Corben

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Gaorav P. Gupta

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Salvatore Piscuoglio

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

A.Y. Ho

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ayca Gucalp

Memorial Sloan Kettering Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge