Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Radiographics ; 44(3): e230136, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38358935

ABSTRACT

The 2021 World Health Organization (WHO) classification system for thoracic tumors (including lung cancer) contains several updates to the 2015 edition. Revisions for lung cancer include a new grading system for invasive nonmucinous adenocarcinoma that better reflects prognosis, reorganization of squamous cell carcinomas and neuroendocrine neoplasms, and description of some new entities. Moreover, remarkable advancements in our knowledge of genetic mutations and targeted therapies have led to a much greater emphasis on genetic testing than that in 2015. In 2015, guidelines recommended evaluation of only two driver mutations, ie, epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) fusions, in patients with nonsquamous non-small cell lung cancer. The 2021 guidelines recommend testing for numerous additional gene mutations for which targeted therapies are now available including ROS1, RET, NTRK1-3, KRAS, BRAF, and MET. The correlation of imaging features and genetic mutations is being studied. Testing for the immune biomarker programmed death ligand 1 is now recommended before starting first-line therapy in patients with metastatic non-small cell lung cancer. Because 70% of lung cancers are unresectable at patient presentation, diagnosis of lung cancer is usually based on small diagnostic samples (ie, biopsy specimens) rather than surgical resection specimens. The 2021 version emphasizes differences in the histopathologic interpretation of small diagnostic samples and resection specimens. Radiologists play a key role not only in evaluation of tumor and metastatic disease but also in identification of optimal biopsy targets. ©RSNA, 2024 Test Your Knowledge questions in the supplemental material and the slide presentation from the RSNA Annual Meeting are available for this article.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/genetics , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , World Health Organization , Molecular Biology
2.
Oncologist ; 28(11): e1065-e1074, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37156009

ABSTRACT

INTRODUCTION: Immune checkpoint inhibitor (ICI) pneumonitis causes substantial morbidity and mortality. Estimates of real-world incidence and reported risk factors vary substantially. METHODS: We conducted a retrospective review of 419 patients with advanced non-small cell lung cancer (NSCLC) who were treated with anti-PD-(L)1 with or without anti-CTLA-4 therapy. Clinical, imaging, and microbiological data were evaluated by multidisciplinary adjudication teams. The primary outcome of interest was grade ≥2 (CTCAEv5) pneumonitis. Clinicopathologic variables, tobacco use, cancer therapies, and preexisting lung disease were assessed for univariate effects using Cox proportional hazards models. We created multivariate Cox proportional hazards models to assess risk factors for pneumonitis and mortality. Pneumonitis, pneumonia, and progression were modeled as time-dependent variables in mortality models. RESULTS: We evaluated 419 patients between 2013 and 2021. The cumulative incidence of pneumonitis was 9.5% (40/419). In a multivariate model, pneumonitis increased the risk for mortality (HR 1.6, 95% CI, 1.0-2.5), after adjustment for disease progression (HR 1.6, 95% CI, 1.4-1.8) and baseline shortness of breath (HR 1.5, 95% CI, 1.2-2.0). Incomplete resolution was more common with more severe pneumonitis. Interstitial lung disease was associated with higher risk for pneumonitis (HR 5.4, 95% CI, 1.1-26.6), particularly in never smokers (HR 26.9, 95% CI, 2.8-259.0). CONCLUSION: Pneumonitis occurred at a high rate and significantly increased mortality. Interstitial lung disease, particularly in never smokers, increased the risk for pneumonitis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Diseases, Interstitial , Lung Neoplasms , Pneumonia , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Incidence , Lung Neoplasms/drug therapy , Pneumonia/epidemiology , Risk Factors , Lung Diseases, Interstitial/complications , Retrospective Studies
3.
Adv Anat Pathol ; 30(4): 280-291, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-36395181

ABSTRACT

Malignant mesothelioma is a rare tumor arising from the mesothelial cells that line the pleura, pericardium, peritoneum, and tunica vaginalis. Imaging plays a primary role in the diagnosis, staging, and management of malignant mesothelioma. Multimodality imaging, including radiography, computed tomography (CT), magnetic resonance imaging (MRI), and F-18 fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT), is used in a variety of scenarios, including diagnosis, guidance for tissue sampling, staging, and reassessment of disease after therapy. CT is the primary imaging modality used in staging. MRI has superior contrast resolution compared with CT and can add value in terms of determining surgical resectability in equivocal cases. MRI can further assess the degree of local invasion, particularly into the mediastinum, chest wall, and diaphragm, for malignant pleural and pericardial mesotheliomas. FDG PET/CT plays a role in the diagnosis and staging of malignant pleural mesothelioma (MPM) and has been shown to be more accurate than CT, MRI, and PET alone in the staging of malignant pleural mesothelioma. PET/CT can also be used to target lesions for biopsy and to assess prognosis, treatment response, and tumor recurrence.


Subject(s)
Mesothelioma, Malignant , Mesothelioma , Pleural Neoplasms , Humans , Mesothelioma, Malignant/pathology , Positron Emission Tomography Computed Tomography/methods , Fluorodeoxyglucose F18 , Pleura/pathology , Pleural Neoplasms/diagnostic imaging , Pleural Neoplasms/pathology , Neoplasm Staging , Mesothelioma/diagnostic imaging , Mesothelioma/pathology , Pericardium/diagnostic imaging , Pericardium/pathology
4.
Cancer ; 128(14): 2736-2745, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35452134

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICI), combined with hypomethylating agents, can be used to treat acute myeloid leukemia (AML), but this strategy results in a high rate of pneumonitis. The authors sought to determine risk factors for pneumonitis development and whether pneumonitis increased mortality. METHODS: The authors conducted a retrospective review of 258 AML patients who received ICI-containing regimens from 2016 to 2018. A multidisciplinary adjudication committee diagnosed pneumonia and pneumonitis by reviewing symptoms, imaging, microbiology, and response to therapies. To measure risk factors for pneumonitis and mortality, multivariate Cox proportional hazards models were constructed. Pneumonia, pneumonitis, and disease progression were modeled as a time-dependent variable and incorporated a standard risk set modifying variables into the models. RESULTS: Thirty patients developed pneumonitis (12%). Of these, 17 had partial or complete resolution, whereas 13 patients died from pneumonitis. Increasing age (hazard ratio [HR], 1.04 per year; 95% confidence interval [CI], 1.00-1.08), and baseline shortness of breath increased pneumonitis risk (HR, 2.51; 95% CI, 1.13-5.55). Female sex (HR, 0.33; 95% CI, 0.15-0.70) and increasing platelet count (HR, 0.52 per log-unit increase; 95% CI, 0.30-0.92) decreased pneumonitis risk. In adjusted models, ICI-related pneumonitis significantly increased mortality (HR, 2.84; 95% CI, 1.84-4.37). CONCLUSIONS: ICI-related pneumonitis occurs at a high rate in AML patients and increases mortality. LAY SUMMARY: Immune checkpoint inhibitors (ICIs) remove inhibitory signals that reduce T-cell function and allow T-cells to better attack cancer cells. In acute myeloid leukemia (AML), the effectiveness of ICIs is limited in part by inflammation of the lung, called pneumonitis. This study reviewed 258 patients with AML who received ICIs and identified 30 patients who developed pneumonitis, nearly half of whom died. Older age and baseline shortness of breath increased pneumonitis risk, whereas female sex and higher baseline platelet counts decreased pneumonitis risk. Pneumonitis increased mortality by nearly 3-fold. This work highlights the significant harm imposed by pneumonitis after ICI therapies.


Subject(s)
Antineoplastic Agents, Immunological , Leukemia, Myeloid, Acute , Lung Neoplasms , Pneumonia , Antineoplastic Agents, Immunological/therapeutic use , Dyspnea/chemically induced , Female , Humans , Immune Checkpoint Inhibitors/adverse effects , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/drug therapy , Lung Neoplasms/drug therapy , Pneumonia/chemically induced , Pneumonia/diagnosis , Pneumonia/epidemiology , Retrospective Studies
5.
Radiographics ; 42(7): 1956-1974, 2022.
Article in English | MEDLINE | ID: mdl-36240075

ABSTRACT

The normal immune system identifies and eliminates precancerous and cancerous cells. However, tumors can develop immune resistance mechanisms, one of which involves the exploitation of pathways, termed immune checkpoints, that normally suppress T-cell function. The goal of immune checkpoint inhibitor (ICI) immunotherapy is to boost T-cell-mediated immunity to mount a more effective attack on cancer cells. ICIs have changed the treatment landscape of advanced non-small cell lung cancer (NSCLC), and numerous ICIs have now been approved as first-line treatments for NSCLC by the U.S. Food and Drug Administration. ICIs can cause atypical response patterns such as pseudoprogression, whereby the tumor burden initially increases but then decreases. Therefore, response criteria have been developed specifically for patients receiving immunotherapy. Because ICIs activate the immune system, they can lead to inflammatory side effects, termed immune-related adverse events (irAEs). Usually occurring within weeks to months after the start of therapy, irAEs range from asymptomatic abnormal laboratory results to life-threatening conditions such as encephalitis, pneumonitis, myocarditis, hepatitis, and colitis. It is important to be aware of the imaging appearances of the various irAEs to avoid misinterpreting them as metastatic disease, progressive disease, or infection. The basic principles of ICI therapy; indications for ICI therapy in the setting of NSCLC; response assessment and atypical response patterns of ICI therapy, as compared with conventional chemotherapy; and the spectrum of irAEs seen at imaging are reviewed. An invited commentary by Nishino is available online. ©RSNA, 2022.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug-Related Side Effects and Adverse Reactions , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Immune Checkpoint Inhibitors , Lung Neoplasms/pathology , Immunotherapy/adverse effects
6.
Radiographics ; 42(1): 176-194, 2022.
Article in English | MEDLINE | ID: mdl-34990326

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy is a recently approved breakthrough treatment that has become a new paradigm in treatment of recurrent or refractory B-cell lymphomas and pediatric or adult acute lymphoid leukemia. CAR T cells are a type of cellular immunotherapy that artificially enhances T cells to boost eradication of malignancy through activation of the native immune system. The CAR construct is a synthetically created functional cell receptor grafted onto previously harvested patient T cells, which bind to preselected tumor-associated antigens and thereby activate host immune signaling cascades to attack tumor cells. Advantages include a single treatment episode of 2-3 weeks and durable disease elimination, with remission rates of over 80%. Responses to therapy are more rapid than with conventional chemotherapy or immunotherapy, with intervening short-interval edema. CAR T-cell administration is associated with therapy-related toxic effects in a large percentage of patients, notably cytokine release syndrome, immune effect cell-associated neurotoxicity syndrome, and infections related to immunosuppression. Knowledge of the expected evolution of therapy response and potential adverse events in CAR T-cell therapy and correlation with the timeline of treatment are important to optimize patient care. Some toxic effects are radiologically evident, and familiarity with their imaging spectrum is key to avoiding misinterpretation. Other clinical toxic effects may be occult at imaging and are diagnosed on the basis of clinical assessment. Future directions for CAR T-cell therapy include new indications and expanded tumor targets, along with novel ways to capture T-cell activation with imaging. An invited commentary by Ramaiya and Smith is available online. Online supplemental material is available for this article. ©RSNA, 2022.


Subject(s)
Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Cell- and Tissue-Based Therapy , Child , Cytokine Release Syndrome , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Radiologists , Receptors, Chimeric Antigen/therapeutic use
7.
Semin Respir Crit Care Med ; 43(6): 862-873, 2022 12.
Article in English | MEDLINE | ID: mdl-35815631

ABSTRACT

Lung cancer is a leading cause of cancer-related mortality worldwide. Imaging is integral in accurate clinical staging to stratify patients into groups to predict survival and determine treatment. The eighth edition of the tumor, node, and metastasis (TNM-8) staging system proposed by the International Association for the Study of Lung Cancer in 2016, accepted by both the Union for International Cancer Control and the American Joint Committee on Cancer, is the current standard method of staging lung cancer. This single TNM staging is used for all histologic subtypes of lung cancer, including nonsmall cell lung cancer, small cell lung cancer, and bronchopulmonary carcinoid tumor, and it addresses both clinical and pathologic staging. Familiarity with the strengths and limitations of imaging modalities used in staging, the nuances of TNM-8, its correct nomenclature, and potential pitfalls are important to optimize patient care. In this article, we discuss the role of computed tomography (CT) and positron emission tomography/CT in lung cancer staging, as well as current imaging recommendations pertaining to TNM-8.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Neoplasm Staging , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/pathology , Small Cell Lung Carcinoma/pathology , Lung/pathology , Prognosis
8.
Semin Diagn Pathol ; 39(2): 92-98, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34167848

ABSTRACT

In the imaging of the mediastinum, benign lesions mimicking malignancy constitute potential pitfalls in interpretation. Localization and characteristic imaging features are key to narrow the differential diagnosis and avoid potential pitfalls in interpretation. Based on certain anatomic landmarks, the mediastinal compartment model enables accurate localization. Depending on the anatomic origin, mediastinal lesions can have various etiologies. The anatomic location and structures contained within each mediastinal compartment are helpful in generating the differential diagnoses. These structures include thyroid, thymus, parathyroid, lymph nodes, pericardium, embryogenic remnants, and parts of the enteric tracts, vessels, and nerves. Imaging characteristics on computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography-computed tomography (PET/CT), including attenuation (fluid, fat, calcification), contrast enhancement, and metabolic activity, aid in narrowing the differential diagnoses. Understanding the roles and limitations of various imaging modalities is helpful in the evaluation of mediastinal masses. In this review, we present potential pitfalls in the imaging of mediastinal lesions with emphasis on the mimics of malignancy.


Subject(s)
Mediastinal Neoplasms , Mediastinum , Humans , Magnetic Resonance Imaging , Mediastinal Neoplasms/diagnostic imaging , Mediastinum/diagnostic imaging , Positron Emission Tomography Computed Tomography , Tomography, X-Ray Computed
9.
J Cardiovasc Nurs ; 36(6): 589-594, 2021.
Article in English | MEDLINE | ID: mdl-34608886

ABSTRACT

BACKGROUND: The patient experience of heart failure involves a multi-impact symptom response with functional limitations, psychological changes, and significant treatment burden. OBJECTIVE: The aim of this study was to examine the change in patient-reported outcomes in newly diagnosed patients with heart failure and reduced ejection fraction (HFrEF) prescribed a wearable cardioverter defibrillator. METHODS: Adults hospitalized for new-onset heart failure, due to ischemic or nonischemic cardiomyopathy, and prescribed a wearable cardioverter defibrillator within 10 days post discharge were approached for inclusion. Participants completed the Kansas City Cardiomyopathy Questionnaire at 3 time points: baseline, day 90, and day 180. RESULTS: A total of 210 patients (26% female) were included. All Kansas City Cardiomyopathy Questionnaire subscales (physical limitation, symptom frequency, quality of life, and social limitation) showed improvement from baseline to day 90 (all Ps < .001). Only quality of life continued to improve from day 90 to day 180 (P < .001). By day 90, nearly 70% of patients showed an improvement in quality of life (67.9%, n = 91), and by day 180, more than 80% (82.8%, n = 111) reported a net improvement. Five patients (3.7%) reported a net decrease, and 18 patients (13.4%) had no net change in quality of life during the 180-day period. CONCLUSION: Patient-reported quality of life improved significantly among patients newly diagnosed with HFrEF and prescribed a wearable cardioverter defibrillator. These results suggest that pursuing guideline-directed medical therapy for HFrEF, while being protected by the wearable cardioverter defibrillator, is likely to provide symptom relief and improve quality of life.


Subject(s)
Heart Failure , Wearable Electronic Devices , Aftercare , Defibrillators , Female , Heart Failure/therapy , Humans , Male , Patient Discharge , Prescriptions , Quality of Life , Stroke Volume
10.
AJR Am J Roentgenol ; 215(6): 1329-1334, 2020 12.
Article in English | MEDLINE | ID: mdl-33021830

ABSTRACT

OBJECTIVE. The purpose of this study is to evaluate the CT and clinical characteristics of in situ pulmonary artery thrombosis (PAT) associated with radiation therapy (RT). MATERIALS AND METHODS. A database search was performed to identify patients who had PAT develop after receiving RT. The CT characteristics of PAT, including the number, location, and appearance of filling defects as well as the presence of associated lung fibrosis, were recorded. The terminology (in situ thrombosis vs acute or chronic pulmonary embolism) used by the interpreting radiologists to describe PAT, the time between the completion of RT and development of PAT, the change in the size of the PAT, and observation of any new thrombi and emboli on follow-up imaging, were also recorded. RESULTS. Of the 27 patients in the study cohort, 22 (81%) had lung cancer and five (19%) had mesothelioma. Most PATs were solitary (93%) and nonocclusive (96%) and formed an obtuse angle to the vessel wall (89%). All PATs were eccentric within the involved PA and were located within the RT volume. The time from completion of RT to initial diagnosis of PAT on CT ranged from 53 to 2522 days (mean, 675 days). Radiation-induced lung fibrosis was present in the ipsilateral lung in all patients. No evidence of additional PA filling defects that suggested embolization were seen on follow-up images of any of the patients, even those who did not receive anticoagulant therapy. CONCLUSION. In situ PAT associated with RT, which to our knowledge has not previously been described in the English literature, has imaging features different from those of acute pulmonary emboli and does not appear to embolize. Radiologist awareness of PAT can facilitate accurate diagnosis and impact management.


Subject(s)
Lung Neoplasms/radiotherapy , Pulmonary Artery , Pulmonary Embolism/diagnostic imaging , Pulmonary Embolism/etiology , Thrombosis/diagnostic imaging , Thrombosis/etiology , Tomography, X-Ray Computed , Adult , Aged , Female , Humans , Male , Middle Aged
11.
Invest New Drugs ; 37(2): 345-351, 2019 04.
Article in English | MEDLINE | ID: mdl-30610588

ABSTRACT

Background Increased adiposity is thought to result in worse clinical outcomes in patients with breast cancer through increased estrogen production, hyperinsulinemia, insulin resistance, and activation of the phosphatidylinositol-3-kinase/AKT/mammalian target of rapamycin (mTOR) pathway. Thus, we hypothesized that the addition of metformin to everolimus and exemestane, could lead to better outcomes in overweight and obese patients with metastatic, hormone receptor-positive, HER2-negative breast cancer. We conducted a phase II trial to evaluate the efficacy and safety of the combination of metformin, everolimus and exemestane in overweight and obese postmenopausal women with metastatic, hormone receptor-positive, HER2-negative breast cancer. Methods Twenty-two patients with a body mass index ≥25 kg/m2 were treated with metformin 1000 mg twice daily, everolimus 10 mg daily and exemestane 25 mg daily. Median progression-free (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. Results Median PFS and OS were 6.3 months (95% confidence interval [CI]: 3.8-11.3 months) and 28.8 months (95% CI: 17.5-59.7 months), respectively. Five patients had a partial response and 7 had stable disease for ≥24 weeks yielding a clinical benefit rate of 54.5%. Compared with overweight patients, obese patients had an improved PFS on univariable (p = 0.015) but not multivariable analysis (p = 0.215). Thirty-two percent of patients experienced a grade 3 treatment-related adverse event (TRAE). There were no grade 4 TRAEs and 7 patients experienced a grade 3 TRAE. Conclusion The combination of metformin, everolimus and exemestane was safe and had moderate clinical benefit in overweight and obese with patients metastatic, hormone receptor-positive, HER2-negative breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Metformin/therapeutic use , Obesity/physiopathology , Overweight/physiopathology , Postmenopause , Adult , Aged , Androstadienes/administration & dosage , Bone Neoplasms/secondary , Breast Neoplasms/pathology , Drug Therapy, Combination , Everolimus/administration & dosage , Female , Follow-Up Studies , Humans , Hypoglycemic Agents/therapeutic use , Middle Aged , Prognosis , Prospective Studies , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Survival Rate
12.
Radiographics ; 39(2): 344-366, 2019.
Article in English | MEDLINE | ID: mdl-30844346

ABSTRACT

Radiation therapy is one of the cornerstones for the treatment of thoracic malignancies. Although advances in radiation therapy technology have improved the delivery of radiation considerably, adverse effects are still common. Postirradiation changes affect the organ or tissue treated and the neighboring structures. Advances in external-beam radiation delivery techniques and how these techniques affect the expected thoracic radiation-induced changes are described. In addition, how to distinguish these expected changes from complications such as infection and radiation-induced malignancy, and identify treatment failure, that is, local tumor recurrence, is reviewed. ©RSNA, 2019.


Subject(s)
Breast/radiation effects , Heart/radiation effects , Lung/drug effects , Neoplasms, Radiation-Induced/diagnostic imaging , Radiation Injuries/diagnostic imaging , Radiography/methods , Radiotherapy/adverse effects , Bone and Bones/diagnostic imaging , Bone and Bones/radiation effects , Breast/diagnostic imaging , Female , Heart/diagnostic imaging , Humans , Lung/diagnostic imaging , Lymph Nodes/diagnostic imaging , Lymph Nodes/radiation effects , Male , Radiation Dosage , Radiation Injuries/etiology , Radiotherapy/methods , Tomography, X-Ray Computed
13.
Radiographics ; 39(1): 44-61, 2019.
Article in English | MEDLINE | ID: mdl-30620703

ABSTRACT

Leukemias are malignancies in which abnormal white blood cells are produced in the bone marrow, resulting in compromise of normal bone marrow hematopoiesis and subsequent cytopenias. Leukemias are classified as myeloid or lymphoid depending on the type of abnormal cells produced and as acute or chronic according to cellular maturity. The four major types of leukemia are acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, and chronic lymphocytic leukemia. Clinical manifestations are due to either bone marrow suppression (anemia, thrombocytopenia, or neutropenia) or leukemic organ infiltration. Imaging manifestations of leukemia in the thorax are myriad. While lymphadenopathy is the most common manifestation of intrathoracic leukemia, leukemia may also involve the lungs, pleura, heart, and bones and soft tissues. Myeloid sarcomas occur in 5%-7% of patients with acute myeloid leukemia and represent masses of myeloid blast cells in an extramedullary location. ©RSNA, 2019.


Subject(s)
Leukemia, Lymphoid/diagnostic imaging , Leukemia, Myeloid/diagnostic imaging , Radiography, Thoracic , Thorax/diagnostic imaging , Diagnosis, Differential , Female , Humans , Leukemia, Lymphoid/pathology , Leukemia, Myeloid/pathology , Male , Positron-Emission Tomography , Risk Factors , Tomography, X-Ray Computed
14.
Invest New Drugs ; 36(2): 299-306, 2018 04.
Article in English | MEDLINE | ID: mdl-29374384

ABSTRACT

Background Tipifarnib is an orally active, competitive inhibitor of farnesyltransferase which has shown encouraging signs of activity either alone or when combined with other agents. Clinical studies of tipifarnib in combination with anti-estrogen therapy have yielded disappointing results. In contrast, tipifarnib appears to be synergistic in combination with anthracycline based chemotherapy. Here we report the results of the first prospective phase II trial evaluating the efficacy of the novel combination of tipifarnib and gemcitabine in the treatment of metastatic breast cancer. Patients and Methods 30 postmenopausal women with metastatic breast cancer were treated on a 21-day cycle with tipifarnib 300 mg PO twice daily from days 1 through 14. Gemcitabine was administered intravenously at a dose of 1000 mg/m2 on days 1 and 8. Patients were treated until disease progression or unacceptable toxicity. Results There was one complete response and four partial responses yielding an objective response rate of 16.7%. Median progression-free survival and overall survival was 2.5 months (95% confidence interval: 1.6-5.7 months) and 13.1 months (95% confidence interval: 9.1-20.6 months), respectively. 40% of patients experienced grade 4 neutropenia in this study. Conclusion The combination of tipifarnib and gemcitabine is not well tolerated with high rates of myelosuppression and is not more effective than gemcitabine monotherapy in the treatment of metastatic breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Deoxycytidine/analogs & derivatives , Quinolones/therapeutic use , Adult , Aged , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Metastasis , Quinolones/adverse effects , Treatment Outcome , Gemcitabine
15.
Radiographics ; 37(1): 52-72, 2017.
Article in English | MEDLINE | ID: mdl-28076011

ABSTRACT

Tuberculosis is a public health problem worldwide, including in the United States-particularly among immunocompromised patients and other high-risk groups. Tuberculosis manifests in active and latent forms. Active disease can occur as primary tuberculosis, developing shortly after infection, or postprimary tuberculosis, developing after a long period of latent infection. Primary tuberculosis occurs most commonly in children and immunocompromised patients, who present with lymphadenopathy, pulmonary consolidation, and pleural effusion. Postprimary tuberculosis may manifest with cavities, consolidations, and centrilobular nodules. Miliary tuberculosis refers to hematogenously disseminated disease that is more commonly seen in immunocompromised patients, who present with miliary lung nodules and multiorgan involvement. The principal means of testing for active tuberculosis is sputum analysis, including smear, culture, and nucleic acid amplification testing. Imaging findings, particularly the presence of cavitation, can affect treatment decisions, such as the duration of therapy. Latent tuberculosis is an asymptomatic infection that can lead to postprimary tuberculosis in the future. Patients who are suspected of having latent tuberculosis may undergo targeted testing with a tuberculin skin test or interferon-γ release assay. Chest radiographs are used to stratify for risk and to assess for asymptomatic active disease. Sequelae of previous tuberculosis that is now inactive manifest characteristically as fibronodular opacities in the apical and upper lung zones. Stability of radiographic findings for 6 months distinguishes inactive from active disease. Nontuberculous mycobacterial disease can sometimes mimic the findings of active tuberculosis, and laboratory confirmation is required to make the distinction. Familiarity with the imaging, clinical, and laboratory features of tuberculosis is important for diagnosis and management. ©RSNA, 2017.


Subject(s)
Bacterial Typing Techniques/methods , Mycobacterium tuberculosis/isolation & purification , Radiography, Thoracic/methods , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/therapy , Diagnosis, Differential , Humans , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/genetics , Tuberculosis, Pulmonary/microbiology
16.
J Comput Assist Tomogr ; 40(6): 917-922, 2016.
Article in English | MEDLINE | ID: mdl-27454787

ABSTRACT

OBJECTIVE: The purpose of this study was to report the computed tomography (CT) findings of non-pneumophila Legionella pneumonia and to compare these CT findings to those caused by Legionella pneumophila in oncologic patients. METHODS: Chest CT scans of 34 oncologic patients with culture-proven Legionella infection (16 L. pneumophila and 18 non-pneumophila Legionella) were retrospectively reviewed. Radiologic checkpoints included consolidation, ground-glass opacities, cavitation, nodules, tree-in-bud opacities, septal thickening, pleural effusions, and adenopathy, as well as the halo, reversed halo, and bulging fissure signs. RESULTS: The most common imaging feature of Legionella pneumonia was consolidation, seen in 94% of patients. Ground-glass opacities were the next most common abnormality. The halo sign was present in 26% of patients, in both immunocompetent and immunosuppressed hosts. Most features occurred with similar frequency between L. pneumophila and non-pneumophila Legionella. CONCLUSIONS: Findings in L. pneumophila pneumonia and non-pneumophila Legionella pneumonia are similar but nonspecific. Airspace consolidation is almost always present; the halo sign is not uncommon.


Subject(s)
Legionellosis/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Pneumonia, Bacterial/diagnostic imaging , Radiography, Thoracic/methods , Tomography, X-Ray Computed/methods , Adult , Aged , Aged, 80 and over , Female , Humans , Legionella/isolation & purification , Legionellosis/microbiology , Lung Neoplasms/microbiology , Male , Middle Aged , Pneumonia, Bacterial/microbiology , Reproducibility of Results , Sensitivity and Specificity
17.
Clin Radiol ; 69(3): 307-14, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24290055

ABSTRACT

In oncological imaging, staging with computed tomography (CT) is widely used to determine treatment. Misinterpretation of fluid in pericardial recesses as mediastinal adenopathy can lead to inaccurate clinical staging and inappropriate management. In this review, we describe normal pericardial anatomy and illustrate imaging features to differentiate fluid in pericardial sinuses and recesses from mediastinal adenopathy.


Subject(s)
Lymphatic Diseases/diagnostic imaging , Mediastinum/diagnostic imaging , Neoplasms/pathology , Pericardial Effusion/diagnostic imaging , Pericardium/diagnostic imaging , Tomography, X-Ray Computed , Contrast Media , Diagnosis, Differential , Humans , Lymphatic Diseases/pathology , Mediastinum/pathology , Pericardial Effusion/pathology , Pericardium/pathology
18.
Clin Chest Med ; 45(2): 325-337, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816091

ABSTRACT

Drug-induced lung disease is commonly encountered, especially in the oncology setting. Diagnosis is challenging because clinical and radiologic findings are nonspecific, often overlapping with other lung pathologies in these patients due to underlying neoplasia, infection, or other treatment effects such as radiotherapy. Furthermore, oncology patients often receive multiple antineoplastic agents concurrently, and virtually every agent has an association with lung injury. In this article, we will review a variety of antineoplastic agents that are associated with drug-induced injury and discuss incidence, their typical timing of onset, and imaging features.


Subject(s)
Antineoplastic Agents , Immunotherapy , Humans , Antineoplastic Agents/adverse effects , Immunotherapy/adverse effects , Lung Diseases/chemically induced , Lung Diseases/etiology , Neoplasms/drug therapy , Neoplasms/complications
19.
Clin Chest Med ; 45(2): 295-305, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816089

ABSTRACT

Lung cancer remains one of the leading causes of mortality worldwide, as well as in the United States. Clinical staging, primarily with imaging, is integral to stratify patients into groups that determine treatment options and predict survival. The eighth edition of the tumor, node, metastasis (TNM-8) staging system proposed in 2016 by the International Association for the Study of Lung Cancer remains the current standard for lung cancer staging. The system is used for all subtypes of lung cancer, including non-small cell lung cancer, small cell lung cancer, and bronchopulmonary carcinoid tumors.


Subject(s)
Lung Neoplasms , Neoplasm Staging , Humans , Lung Neoplasms/pathology , Lung Neoplasms/diagnostic imaging , Neoplasm Staging/methods , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Tomography, X-Ray Computed , Diagnostic Imaging/methods , Positron-Emission Tomography
20.
Clin Chest Med ; 45(2): 237-248, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38816085

ABSTRACT

The pericardium comprises a double-walled fibrous-serosal sac that encloses the heart. Reflections of the serosal layer form sinuses and recesses. With advances in multidetector computed tomography (CT) technology, pericardial recesses are frequently detected with thin-section CT. Knowledge of pericardial anatomy on imaging is crucial to avoid misinterpretation of fluid-filled pericardial sinuses and recesses as adenopathy/pericardial metastasis or aortic dissection, which can impact patient management and treatment decisions. The authors offer a comprehensive review of pericardial anatomy and its variations observed on CT, potential pitfalls in image interpretation, and implications for the pulmonologist with respect to unnecessary diagnostic procedures or interventions.


Subject(s)
Pericardium , Humans , Pericardium/diagnostic imaging , Tomography, X-Ray Computed , Pulmonologists , Multidetector Computed Tomography/methods
SELECTION OF CITATIONS
SEARCH DETAIL