Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Open Forum Infect Dis ; 11(1): ofad632, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38264094

ABSTRACT

Mpox, caused by infection with Monkeypox virus, usually presents as a mild, self-limited illness in immunocompetent persons that resolves within 2-4 weeks. Serious complications have been reported when mpox lesions involve vulnerable anatomic sites, such as the eye, and in those with substantial immunosuppression. We describe a patient with advanced human immunodeficiency virus infection and sustained viral shedding of mpox with ocular involvement, which resulted in vision loss.

2.
JAMA Ophthalmol ; 142(2): 157-158, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38206632

ABSTRACT

This case report describes a conjunctival biopsy of a patient with mpox infection.


Subject(s)
Mpox (monkeypox) , Humans , Biopsy , Conjunctiva
3.
Mayo Clin Proc Innov Qual Outcomes ; 7(5): 437-442, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37752967

ABSTRACT

Supernumerary nipples develop on the chest and abdominopelvic regions along the embryonic milk line. Their anatomy varies from isolated accessory nipples to complete supernumerary nipples (accessory nipple, areola, and underlying glandular breast tissue). Patients with a pathogenic BReast CAncer (BRCA) sequence variation are at an increased cumulative risk of developing breast cancer, and it is the standard of care for them to be offered medical or surgical risk reduction. Given the relatively low prevalence of breast cancer within supernumerary nipples and ectopic glandular breast tissue, no current recommendations exist to guide multidisciplinary management of patients with BRCA sequence variations and ectopic breast tissue. Our case is of a 62-year-old female BRCA-1 carrier with a previous history of right breast cancer who developed a new primary breast cancer within a supernumerary nipple after undergoing surgical risk reduction. With no current consensus on the surgical management of supernumerary nipples in BRCA-1 carriers, our recommendation is to perform a thorough physical examination before risk-reducing operation. If supernumerary nipples or ectopic glandular breast tissue are present, wide-local excision of the tissue should be offered for more complete surgical risk reduction.

5.
Clin Breast Cancer ; 22(2): 186-190, 2022 02.
Article in English | MEDLINE | ID: mdl-34462208

ABSTRACT

BACKGROUND: Neoadjuvant therapy aims to preoperatively downstage breast cancer patients. We evaluated nodal upstaging in clinically node-negative (cN0) patients receiving neoadjuvant chemotherapy (NAC) and neoadjuvant endocrine therapy (NET). METHODS: cN0 patients undergoing neoadjuvant therapy from 2009 to 2018 were reviewed. Univariate and multivariate analyses evaluated rates of nodal upstaging. RESULTS: A total of 228 cN0 patients with a mean age of 55 years underwent neoadjuvant therapy for Stage I-III invasive carcinoma. Subtypes included ER+/HER2- = 93 (40%), HER2+ = 61 (27%), and triple negative (TNBC) = 74 (33%). Among ER+/HER2- patients, 65 (70%) underwent NET. Overall, 49 patients (21%) were upstaged due to occult nodal disease. Factors associated with higher rates of occult nodal disease included advanced stage on initial presentation (P = .008), larger presenting tumor size (P = .009), low/intermediate tumor grade (P = .025), and ER+/HER2- subtype (P < .001); incidence of occult nodal disease by subtype included: ER+/HER2- = 37%, HER2+ = 15%, TNBC = 8%. Patients experiencing a breast pCR had a significantly lower rate of nodal upstaging compared to those with residual tumor (4% vs. 96%, P < .001). On multivariate analysis, ER+/HER- patients exhibited higher risk of occult nodal disease when compared to patients with HER2+ (odds ratio [OR] = 3.4, 95% CI, 1.2-9.8, P = .003) and TNBC (OR = 5.7, 95% CI, 1.7-19.6, P = .003). Comparing NAC vs. NET in ER+/HER2- patients showed no difference in rates of occult nodal disease (39% vs. 35%, P = .13). CONCLUSIONS: ER+/HER2- subtype carries higher risk for occult nodal disease after neoadjuvant therapy; NAC versus NET in these patients does not affect nodal upstaging.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Neoplasm, Residual/pathology , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Adult , Aged , Breast Neoplasms/metabolism , Female , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neoadjuvant Therapy , Neoplasm Invasiveness
6.
Sci Rep ; 11(1): 10571, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011996

ABSTRACT

Breast cancers exhibit intratumoral heterogeneity associated with disease progression and therapeutic resistance. To define the sources and the extent of heterogeneity, we performed an in-depth analysis of the genomic architecture of three chemoradiation-naïve breast cancers with well-defined clinical features including variable ER, PR, ERBB2 receptor expression and two distinct pathogenic BRCA2mut genotypes. The latter included a germ line carrier and a patient with a somatic variant. In each case we combined DNA content-based flow cytometry with whole exome sequencing and genome wide copy number variant (CNV) analysis of distinct populations sorted from multiple (4-18) mapped biopsies within the tumors and involved lymph nodes. Interrogating flow-sorted tumor populations from each biopsy provided an objective method to distinguish fixed and variable genomic lesions in each tumor. Notably we show that tumors exploit CNVs to fix mutations and deletions in distinct populations throughout each tumor. The identification of fixed genomic lesions that are shared or unique within each tumor, has broad implications for the study of tumor heterogeneity including the presence of tumor markers and therapeutic targets, and of candidate neoepitopes in breast and other solid tumors that can advance more effective treatment and clinical management of patients with disease.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Evolution, Molecular , Aneuploidy , Biomarkers, Tumor , Humans
7.
Eur J Cancer ; 148: 287-296, 2021 05.
Article in English | MEDLINE | ID: mdl-33765513

ABSTRACT

AIM: To present the pre-specified analyses of >5-years follow-up of the Phase III ALTTO trial. PATIENTS AND METHODS: 8381 patients with stage I-III HER2 positive breast cancer randomised to chemotherapy plus 1-year of trastuzumab (T), oral lapatinib (L; no longer evaluated), trastuzumab followed by lapatinib (T→L), and lapatinib + trastuzumab (L+T). The primary endpoint was disease-free survival (DFS). A secondary analysis examined DFS treatment effects by hormone receptor status, nodal status and chemotherapy timing; time to recurrence; overall survival (OS) and safety (overall and cardiac). RESULTS: At a median follow-up of 6.9 years, 705 DFS events for L+T versus T were observed. Hazard Ratio (HR) for DFS was 0.86 (95% CI, 0.74-1.00) for L+T versus T and 0.93 (95% CI, 0.81-1.08) for T→L versus T. The 6-year DFS were 85%, 84%, and 82% for L+T, T→L, and T, respectively. HR for OS was 0.86 (95% CI, 0.70-1.06) for L+T versus T and 0.88 (95% CI, 0.71-1.08) for T→L versus T. The 6-year OS were 93%, 92%, and 91% for L+T, T→L, and T, respectively. Subset analyses showed a numerically better HR for DFS in favour of L+T versus T for the hormone-receptor-negative [HR 0.80 (95% CI, 0.64-1.00; 6-yr DFS% = 84% versus 80%)] and the sequential chemotherapy [HR 0.83 (95% CI, 0.69-1.00; 6-yr DFS% = 83% versus79%)] subgroups. CONCLUSION: T+L did not significantly improve DFS and OS over T alone, both with chemotherapy, and, therefore, cannot be recommended for adjuvant treatment of early-stage HER2-positive breast cancer. TRIAL REGISTRATION: clinicaltrials.gov Identifier NCT00490139.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Chemotherapy, Adjuvant/mortality , Neoadjuvant Therapy/mortality , Adolescent , Adult , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Prospective Studies , Survival Rate , Young Adult
9.
World Neurosurg ; 145: 340-347, 2021 01.
Article in English | MEDLINE | ID: mdl-32987172

ABSTRACT

BACKGROUND: A collision tumor is a rare entity consisting of 2 histologically distinct tumor types (benign or malignant) in the same anatomic location. This can occur from a tumor-to-tumor metastasis or as a result of 2 adjacent intracranial tumors colliding and growing together. To our knowledge, this is the first reported case of collision tumor with confirmed meningioma and uterine adenocarcinoma. Multiple mechanisms have been proposed for the facilitative growth of collision tumors, including local epigenetic signaling. Clinically, it is important to consider collision tumors in the differential diagnosis of a rapidly growing intracranial lesion in the setting of systemic cancer to provide optimal surgical and postoperative management. CASE DESCRIPTION: A 78-year-old, right-handed woman with a known 10-year history of stable meningioma presented for evaluation of a right sphenoid wing lesion. She had recently completed treatment of uterine papillary serous carcinoma with no evidence of disease on follow-up imaging. On presentation, there was significant progression of the meningioma resulting in brain compression and right third nerve palsy. The patient underwent urgent resection of the lesion. Pathology demonstrated a collision tumor with a combination of metastatic uterine papillary serous carcinoma and meningioma. CONCLUSIONS: It is important to consider a collision tumor when a patient with a benign intracranial lesion presents with rapid progression, even in the context of a systemic cancer that rarely metastasizes to the brain. Appropriate histopathologic assessment is crucial in these cases and can have a significant impact on treatment plan and prognosis.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/secondary , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Meningioma/pathology , Uterine Neoplasms/pathology , Adenocarcinoma/surgery , Aged , Brain Neoplasms/surgery , Disease Progression , Fatal Outcome , Female , Humans , Magnetic Resonance Imaging , Meningioma/complications , Meningioma/surgery , Nerve Compression Syndromes/etiology , Nerve Compression Syndromes/pathology , Oculomotor Nerve/pathology , Oculomotor Nerve Diseases/pathology , Uterine Neoplasms/surgery
10.
J Surg Oncol ; 122(4): 619-622, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32506815

ABSTRACT

BACKGROUND AND OBJECTIVES: Neoadjuvant endocrine therapy (NET) for ER+ breast cancer can downstage primary tumors. We evaluated NET efficacy in node-positive patients. METHODS: Node-positive patients undergoing NET for ER+ breast cancer from 2012 to 2019 were reviewed. Primary endpoints included rates of axillary lymphadenectomy (ALND), pathologic complete response (pCR), and final nodal staging. RESULTS: Thirty-nine patients were included. Before NET, all were clinically node-positive (cN1 = 36, 94%; cN2 = 2, 5%; cN3 = 1, 3%; Stage II = 23, 59%, Stage III = 16, 41%). After NET, nine (23%) had clinically persistent axillary disease necessitating ALND. The remaining 30 (77%) underwent sentinel lymph node biopsy (SLNB). Of these, 25 (83%) were SLNB+ on frozen section, undergoing immediate ALND. Five patients were negative on frozen section: one had a confirmed axillary pCR, and four had residual nodal disease on permanent pathology. One underwent delayed ALND, and for the remaining three patients, decision was made to forgo ALND. Final overall axillary staging was: N0 (pCR) = 1, 3%, pN1mic = 1, 3%, pN1 = 20, 51%, pN2 = 12, 30%, pN3 = 5, 13%; Stage II = 16, 41%, Stage III = 23, 59%. CONCLUSIONS: While NET is reported to downstage primary tumors, downstaging of the axilla was unsuccessful in the majority of patients.

11.
J Clin Invest ; 130(4): 1931-1947, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31917686

ABSTRACT

Visceral adipose tissue plays a critical role in numerous diseases. Although imaging studies often show adipose involvement in abdominal diseases, their outcomes may vary from being a mild self-limited illness to one with systemic inflammation and organ failure. We therefore compared the pattern of visceral adipose injury during acute pancreatitis and acute diverticulitis to determine its role in organ failure. Acute pancreatitis-associated adipose tissue had ongoing lipolysis in the absence of adipocyte triglyceride lipase (ATGL). Pancreatic lipase injected into mouse visceral adipose tissue hydrolyzed adipose triglyceride and generated excess nonesterified fatty acids (NEFAs), which caused organ failure in the absence of acute pancreatitis. Pancreatic triglyceride lipase (PNLIP) increased in adipose tissue during pancreatitis and entered adipocytes by multiple mechanisms, hydrolyzing adipose triglyceride and generating excess NEFAs. During pancreatitis, obese PNLIP-knockout mice, unlike obese adipocyte-specific ATGL knockouts, had lower visceral adipose tissue lipolysis, milder inflammation, less severe organ failure, and improved survival. PNLIP-knockout mice, unlike ATGL knockouts, were protected from adipocyte-induced pancreatic acinar injury without affecting NEFA signaling or acute pancreatitis induction. Therefore, during pancreatitis, unlike diverticulitis, PNLIP leaking into visceral adipose tissue can cause excessive visceral adipose tissue lipolysis independently of adipocyte-autonomous ATGL, and thereby worsen organ failure.


Subject(s)
Adipocytes/enzymology , Intra-Abdominal Fat/enzymology , Lipase/metabolism , Pancreatitis/enzymology , Signal Transduction , Acute Disease , Adipocytes/pathology , Animals , Fatty Acids, Nonesterified/genetics , Fatty Acids, Nonesterified/metabolism , Female , Humans , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Intra-Abdominal Fat/pathology , Lipase/genetics , Male , Mice , Mice, Knockout , Pancreatitis/genetics , Pancreatitis/pathology
12.
Sci Transl Med ; 11(504)2019 08 07.
Article in English | MEDLINE | ID: mdl-31391323

ABSTRACT

Longitudinal analysis of circulating tumor DNA (ctDNA) has shown promise for monitoring treatment response. However, most current methods lack adequate sensitivity for residual disease detection during or after completion of treatment in patients with nonmetastatic cancer. To address this gap and to improve sensitivity for minute quantities of residual tumor DNA in plasma, we have developed targeted digital sequencing (TARDIS) for multiplexed analysis of patient-specific cancer mutations. In reference samples, by simultaneously analyzing 8 to 16 known mutations, TARDIS achieved 91 and 53% sensitivity at mutant allele fractions (AFs) of 3 in 104 and 3 in 105, respectively, with 96% specificity, using input DNA equivalent to a single tube of blood. We successfully analyzed up to 115 mutations per patient in 80 plasma samples from 33 women with stage I to III breast cancer. Before treatment, TARDIS detected ctDNA in all patients with 0.11% median AF. After completion of neoadjuvant therapy, ctDNA concentrations were lower in patients who achieved pathological complete response (pathCR) compared to patients with residual disease (median AFs, 0.003 and 0.017%, respectively, P = 0.0057, AUC = 0.83). In addition, patients with pathCR showed a larger decrease in ctDNA concentrations during neoadjuvant therapy. These results demonstrate high accuracy for assessment of molecular response and residual disease during neoadjuvant therapy using ctDNA analysis. TARDIS has achieved up to 100-fold improvement beyond the current limit of ctDNA detection using clinically relevant blood volumes, demonstrating that personalized ctDNA tracking could enable individualized clinical management of patients with cancer treated with curative intent.


Subject(s)
Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Circulating Tumor DNA/analysis , Neoadjuvant Therapy , Neoplasm, Residual/blood , Neoplasm, Residual/drug therapy , Biological Assay , Breast Neoplasms/genetics , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Female , Humans , Mutation/genetics , Neoplasm Staging , Neoplasm, Residual/genetics , ROC Curve , Reference Standards , Sequence Analysis, DNA
13.
Am J Surg ; 218(6): 1229-1233, 2019 12.
Article in English | MEDLINE | ID: mdl-31421894

ABSTRACT

BACKGROUND: The Choosing Wisely Organization and the American College of Surgeons have issued recommendations for patients >70 with breast cancer involving screening and use of radiation therapy (RT) and sentinel lymph node biopsies (SNLB) in early stage tumors. This study evaluated compliance and implementation of these recommendations. METHODS: A database of patients undergoing breast cancer surgery was retrospectively queried from 2002 to 2017. Patients were divided into cohorts before and after the year of each guideline publication. RESULTS: The rate of presentation on mammography was not different before 2009 (65%) vs. after 2009 (66%). RT was given to 57% of patients with T1 ER + Her2-prior to 2013 vs. 27% after (p=<0.001). SLNB was performed in 91% of patients with T1, grade1/2, ER + Her2-tumors prior to 2016 vs. 56% after (p=<0.001). CONCLUSION: Rates of mammography detected breast cancer have not decreased but adjuvant RT and SLNB are less frequently performed in low risk breast cancer in the elderly.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Practice Guidelines as Topic , Aged , Aged, 80 and over , Female , Humans , Life Expectancy , Mammography , Neoplasm Staging , Retrospective Studies , Sentinel Lymph Node Biopsy , United States
14.
Breast Cancer Res Treat ; 177(1): 103-114, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31134488

ABSTRACT

PURPOSE: Limited evidence exists on the impact of hormone receptor (HR) status to counsel HER2-positive early breast cancer patients receiving adjuvant anti-HER2 therapy. METHODS: ALTTO (BIG 2-06) was an international, intergroup, open-label, randomized phase III trial in HER2-positive early breast cancer patients randomized to receive 1 year of trastuzumab and/or lapatinib. HER2, estrogen and progesterone receptors were centrally tested for all patients. We investigated the impact of HR status on prognosis, risk of disease-free survival (DFS) events over time, patterns of first DFS events, and factors associated with risk of DFS events overall, in years 0-5 and 6-8. RESULTS: Out of 6273 patients included in this analysis, 3603 (57.4%) had HR-positive tumors. Median follow-up was 6.93 years. Five-year and 8-year DFS were 86% and 80% in patients with HR-positive disease, and 83% and 79% in those with HR-negative tumors, respectively. Mean annual hazards of recurrence in years 0-5 were 3% in patients with HR-positive disease and 4% in those with HR-negative tumors, while in years 6-8 they were 3% and 2%, respectively. Distribution of first DFS event in years 6-8 (P = 0.005) and type of first distant recurrence (P < 0.001) were significantly different between the two groups. Risk factors for DFS events overall, in years 0-5, and 6-8 were different in patients with HR-positive and HR-negative tumors. CONCLUSIONS: HER2-positive early breast cancer is characterized by the presence of two diseases with distinct natural history based on HR status requiring the development of different follow-up strategies and future de-escalation and escalation clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Receptor, ErbB-2/metabolism , Receptors, Progesterone/metabolism , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Molecular Targeted Therapy , Proportional Hazards Models , Receptor, ErbB-2/antagonists & inhibitors , Treatment Outcome
15.
Oncologist ; 24(5): 589-e160, 2019 05.
Article in English | MEDLINE | ID: mdl-30679315

ABSTRACT

LESSONS LEARNED: Dual epidermal growth factor receptor (EGFR)-directed therapy with erlotinib and panitumumab in combination with gemcitabine was superior to gemcitabine and erlotinib, but the clinical relevance is uncertain given the limited role of gemcitabine monotherapy.A significantly longer overall survival was observed in patients receiving the dual EGFR-directed therapy.The dual EGFR-directed therapy resulted in increased toxicity. BACKGROUND: Gemcitabine is active in patients with advanced pancreatic adenocarcinoma. The combination of erlotinib, an oral epidermal growth factor receptor (EGFR) inhibitor, and gemcitabine was shown to modestly prolong overall survival when compared with gemcitabine alone. The North Central Cancer Treatment Group (now part of Alliance for Clinical Trials in Oncology) trial N064B compared gemcitabine plus erlotinib versus gemcitabine plus combined EGFR inhibition with erlotinib and panitumumab. METHODS: Eligible patients with metastatic adenocarcinoma of the pancreas were randomized to either gemcitabine 1,000 mg/m2 on days 1, 8, and 15 of a 28-day cycle with erlotinib 100 mg p.o. daily (Arm A) or the same combination with the addition of panitumumab 4 mg/kg on days 1 and 15 of a 28-day cycle (Arm B). The primary endpoint of the trial was overall survival. Secondary endpoints included progression-free survival, the confirmed response rate, and toxicity. Comparison between arms for the primary endpoint was done with a one-sided log-rank test, and a p value less than .20 was considered statistically significant. Response rate comparison was done with Fisher's exact test. All other reported p values are two-sided. RESULTS: A total of 92 patients were randomized, 46 to each arm. The median overall survival was 4.2 months in Arm A and 8.3 months in Arm B (hazard ratio, 0.817; 95% confidence interval [CI], 0.530-1.260; p = .1792). The progression-free survival was 2.0 months in Arm A and 3.6 months in Arm B (hazard ratio, 0.843; 95% CI, 0.555-1.280; p = .4190). A partial confirmed response was seen in 8.7% of patients on Arm A and 6.5% on Arm B (p = .9999). No patients had a complete response. Grade 3 and higher nonhematologic toxicities were more common in patients on Arm B compared with those on Arm A (82.6% vs. 52.2%; p = .0018). CONCLUSION: Dual EGFR-directed therapy resulted in a significant prolongation of overall survival in patients with advanced adenocarcinoma of the pancreas but was associated with substantially increased toxicities. Dual EGFR-directed therapy in combination with gemcitabine alone cannot be recommended for further study, as single-agent gemcitabine is no longer considered an appropriate therapy for otherwise fit patients with metastatic pancreatic cancer.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Erlotinib Hydrochloride/therapeutic use , Pancreatic Neoplasms/drug therapy , Panitumumab/therapeutic use , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Erlotinib Hydrochloride/pharmacology , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Panitumumab/pharmacology , Survival Analysis , Gemcitabine , Pancreatic Neoplasms
16.
Breast Cancer Res ; 20(1): 71, 2018 07 11.
Article in English | MEDLINE | ID: mdl-29996881

ABSTRACT

BACKGROUND: Elevated PD-L1 expression on tumor cells, a context associated with an adaptive immune response, has been linked to the total burden of copy number variants (CNVs) in aneuploid tumors, to microsatellite instability (MSI), and to specific genomic driver lesions, including loss of PTEN, MYC amplification, and activating mutations in driver oncogenes such as KRAS and PIK3CA. Triple-negative breast cancers (TNBCs) typically have high levels of CNVs and diverse driver lesions in their genomes. Thus, there is significant interest in exploiting genomic data to develop predictive immunotherapy biomarkers for patients with TNBC. METHODS: Whole tissue samples from 55 resected TNBCs were screened by immunohistochemistry (IHC) for PD-1 and PD-L1 by using validated antibodies and established scoring methods for staining of tumor and non-tumor cells. In parallel, we interrogated biopsies from each resection with DNA content flow cytometry and sorted the nuclei of diploid, tetraploid, and aneuploid cell populations. CNVs were mapped with CNV oligonucleotide arrays by using purified (>95%) tumor populations. We generated whole exome data for 12 sorted tumor samples to increase the resolution within loci of interest and to incorporate somatic mutations into our genomic signatures. RESULTS AND CONCLUSIONS: PD-L1 staining was detected on tumor cells in 29 out of 54 (54%) evaluable cases and was associated with increased overall survival (P = 0.0024). High levels of PD-1 and PD-L1 (IHC ≥4) were present in 11 out of 54 (20%) and 20 out of 54 (37%) cases with staining of PD-L1 primarily on tumor cells for 17 out of 20 (85%) cases. The latter included tumors with both high (>50) and low (<20) numbers of CNVs. Notably, homozygous deletion of PTEN (n = 6) or activating mutation in PIK3CA (n = 1) was not associated with increased expression of either immune checkpoint activator in TNBC. In contrast, two treatment-naïve cases with EGFR driver amplicons had high PD-L1 tumor staining. High mutational load and predicted neoepitopes were observed in MSI+ and high CNV burden TNBCs but were not associated with high PD-L1 expression on tumor cells. Our results challenge current models of genomic-based immunotherapy signatures yet suggest that discrete genomic lesions may complement existing biomarkers to advance immune checkpoint therapies for patients with TNBC.


Subject(s)
B7-H1 Antigen/genetics , Biomarkers, Tumor/genetics , DNA Copy Number Variations/genetics , Programmed Cell Death 1 Receptor/genetics , Triple Negative Breast Neoplasms/genetics , Aged , Aneuploidy , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating/pathology , Microsatellite Instability , Middle Aged , Mutation , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Triple Negative Breast Neoplasms/pathology , Exome Sequencing
18.
Am J Surg ; 214(6): 1096-1101, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28943062

ABSTRACT

BACKGROUND: The appropriate management of the axilla among women undergoing neoadjuvant therapy is in evolution. METHOD: A retrospective review of a prospective database of women with breast cancer who underwent neoadjuvant systemic therapy using endocrine/chemotherapy (NE/CT) from 2002 to 2015 was performed. RESULTS: We reviewed 253 women: triple negative breast cancer (30%), ER+HER2- (44%) breast cancer and HER2+ (25%) disease. The mean age was 55 years (SD = 12). 197 patients were analyzed based on their axillary disease. 33 patients (35%) who had clinical N1-3 disease prior to neoadjuvant therapy had no axillary metastases at definitive surgery. There were no significant differences in overall survival or local/regional recurrence between patients who underwent axillary lymph node dissection (ALND), sentinel lymph node biopsy (SLNB), or SLNB+ALND (p = 0.05061 and p = 0.33). CONCLUSION: SLNB is a viable technique in patients with breast cancer undergoing NE/CT. Patients with pre-neoadjuvant therapy proven axillary disease may be a candidate for SLNB as opposed to planned ALND with good multidisciplinary review of their response and localization of previously positive lymph nodes.


Subject(s)
Breast Neoplasms/surgery , Neoadjuvant Therapy , Sentinel Lymph Node Biopsy , Adult , Aged , Aged, 80 and over , Axilla , Breast Neoplasms/pathology , Female , Humans , Lymph Node Excision , Middle Aged , Neoplasm Staging , Retrospective Studies , Survival Rate , Treatment Outcome
19.
Mod Pathol ; 30(11): 1516-1526, 2017 11.
Article in English | MEDLINE | ID: mdl-28752839

ABSTRACT

The amplification of chromosome 9p24.1 encoding PD-L1, PD-L2, and JAK2 has been reported in multiple types of cancer and is associated with poor outcome, upregulation of PD-L1, and activation of the JAK/STAT pathway. We have developed a novel fluorescence in situ hybridization assay which combines 3 probes mapping to 9p24.1 with a commercial chromosome 9 centromere (CEN9) probe for detection of the JAK2/9p24.1 amplification. JAK2 fluorescence in situ hybridization was compared with array-based comparative genomic hybridization in 34 samples of triple negative breast cancer tumor. By array-based comparative genomic hybridization, 15 had 9p24.1 copy-number gain (log2ratio>0.3) and 19 were classified as non-gain (log2ratio≤0.3). Copy-number gain was defined as JAK2/CEN9 ratio ≥1.1 or average JAK2 signals≥3.0. Twelve of 15 samples with copy-number gain by array-based comparative genomic hybridization were also detected by fluorescence in situ hybridization. Eighteen of 19 samples classified as copy-number non-gain by array-based comparative genomic hybridization were concordant by array-based comparative genomic hybridization. The sensitivity and specificity of the fluorescence in situ hybridization assay was 80% and 95%, respectively (P=0.02). The sample with the highest level of amplification by array-based comparative genomic hybridization (log2ratio=3.6) also scored highest by fluorescence in situ hybridization (ratio=8.2). There was a correlation between the expression of JAK2 and amplification status (Mean 633 vs 393, P=0.02), and there was a trend of association with PD-L1 RNA expression (Mean 46 vs 22, P=0.11). No significant association was observed between PD-L1 immunohistochemistry expression and copy-number gain status. In summary, the novel array-based comparative genomic hybridization assay for detection of chromosome 9p24.1 strongly correlates with the detection of copy-number gain by array-based comparative genomic hybridization. In triple negative breast cancer, this biomarker may identify a relevant subset of patients for targeted molecular therapies.


Subject(s)
B7-H1 Antigen/genetics , Biomarkers, Tumor/analysis , In Situ Hybridization, Fluorescence/methods , Janus Kinase 2/genetics , Triple Negative Breast Neoplasms/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Female , Gene Amplification , Humans , Middle Aged , Triple Negative Breast Neoplasms/diagnosis
20.
Clin Cancer Res ; 23(15): 4203-4211, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28533226

ABSTRACT

Background: Preclinical evidence indicates that increased insulin-like growth factor receptor-1 (IGF1R) signaling interferes with the action of trastuzumab suggesting a possible mechanism of trastuzumab resistance. Thus, we evaluated IGF1R prevalence, relationship with demographic data, and association with disease-free survival (DFS) of patients randomized to chemotherapy alone (Arm A) or chemotherapy with sequential (Arm B) or concurrent trastuzumab (Arm C) in the prospective phase III HER2+ adjuvant N9831 trial.Experimental Design: IGF1R protein expression was determined in tissue microarray sections (three cores per block; N = 1,197) or in whole tissue sections (WS; N = 537) using IHC (rabbit polyclonal antibody against IGF1R ß-subunit). A tumor was considered positive (IGF1R+) if any core or WS had ≥1+ membrane staining in >0% invasive cells. Median follow-up was 8.5 years.Results: Of 1,734 patients, 708 (41%) had IGF1R+ breast tumors. IGF1R+ was associated with younger age (median 48 vs. 51, P = 0.007), estrogen receptor/progesterone receptor positivity (78% vs. 35%, P < 0.001), nodal positivity (89% vs. 83%, P < 0.001), well/intermediate grade (34% vs. 24%, P < 0.001), tumors ≥2 cm (72% vs. 67%, P = 0.02) but not associated with race or tumor histology. IGF1R did not affect DFS within arms. Between Arms A and C, patients with IGF1R+ and IGF1R- tumors had DFS HRs of 0.48 (P ≤ 0.001) and 0.68 (P = 0.009), respectively (Pinteraction = 0.17). Between Arms A and B, patients with IGF1R+ and IGF1R- tumors had DFS HRs of 0.83 (P = 0.25) and 0.69 (P = 0.01), respectively (Pinteraction = 0.42).Conclusions: In contrast to preclinical studies that suggest a decrease in trastuzumab sensitivity in IGF1R+ tumors, our adjuvant data show benefit of adding trastuzumab for patients with either IGF1R+ and IGF1R- breast tumors. Clin Cancer Res; 23(15); 4203-11. ©2016 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Receptors, Somatomedin/genetics , Trastuzumab/administration & dosage , Adult , Aged , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Middle Aged , Neoplasm Staging , Receptor, ErbB-2/genetics , Receptor, IGF Type 1 , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Trastuzumab/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...