Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Cancers (Basel) ; 16(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38791979

ABSTRACT

The vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are key regulators of blood vessel formation, including in tumors, where their deregulated function can promote the production of aberrant, leaky blood vessels, supporting tumor development. Here we investigated the VEGFR1 ligand VEGF-B, which we demonstrate to be expressed in tumor cells and in tumor stroma and vasculature across a range of tumor types. We examined the anti-VEGF-B-specific monoclonal antibody 2H10 in preclinical xenograft models of breast and colorectal cancer, in comparison with the anti-VEGF-A antibody bevacizumab. Similar to bevacizumab, 2H10 therapy was associated with changes in tumor blood vessels and intra-tumoral diffusion consistent with normalization of the tumor vasculature. Accordingly, treatment resulted in partial inhibition of tumor growth, and significantly improved the response to chemotherapy. Our studies indicate the importance of VEGF-B in tumor growth, and the potential of specific anti-VEGF-B treatment to inhibit tumor development, alone or in combination with established chemotherapies.

2.
Invest New Drugs ; 40(4): 747-755, 2022 08.
Article in English | MEDLINE | ID: mdl-35404015

ABSTRACT

Ephrin type-A 2 (EphA2) is a transmembrane receptor expressed in epithelial cancers. We report on a phase I dose escalation and biodistribution study of DS-8895a, an anti-EphA2 antibody, in patients with advanced EphA2 positive cancers. DS-8895a was administered at 1, 3, 10 or 20 mg/kg every 2 weeks to determine safety, pharmacokinetics and anti-tumor efficacy. All patients underwent 89Zr trace-labelled infusion of DS-8895a (89Zr-DS-8995a) positron emission tomography imaging to determine the biodistribution of DS-8895a, and correlate findings with EphA2 expression, receptor saturation and response. Nine patients were enrolled on study. Of patients enrolled, seven patients received at least one infusion of DS-8895a: four patients received 1 mg/kg dose (Cohort 1) and three patients received 3 mg/kg dose (Cohort 2). Median age was 67.0 years (range 52-81), majority male (71%), and median number of prior systemic therapies was three (range 0-8). The primary cancer diagnosis was colorectal cancer (two patients) and one patient each had gastric, head and neck, high-grade serous adenocarcinoma, lung, and pancreatic cancers. No dose-limiting toxicities or treatment-related adverse events reported. The best response for the patients in Cohort 1 was stable disease and in Cohort 2 was progressive disease. 89Zr-DS-8895a demonstrated no normal tissue uptake and specific low-grade uptake in most tumours. DS-8895a had limited therapeutic efficacy at doses evaluated and 89Zr-DS-8895a demonstrated low tumour uptake. The biodistribution data from this study were key in halting further development of DS-8895a, highlighting the importance of biodistribution studies in drug development. (Trial registration: ClinicalTrials.gov Identifier NCT02252211).


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Agents, Immunological , Neoplasms , Aged , Aged, 80 and over , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/therapeutic use , Ephrin-A2/immunology , Humans , Male , Middle Aged , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Receptor, EphA2/drug effects , Tissue Distribution
3.
EBioMedicine ; 65: 103252, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33640794

ABSTRACT

BACKGROUND: A research priority in finding a cure for HIV is to establish methods to accurately locate and quantify where and how HIV persists in people living with HIV (PLWH) receiving suppressive antiretroviral therapy (ART). Infusing copper-64 (64Cu) radiolabelled broadly neutralising antibodies targeting HIV envelope (Env) with CT scan and positron emission tomography (PET) identified HIV Env in tissues in SIV infected non-human primates . We aimed to determine if a similar approach was effective in people living with HIV (PLWH). METHODS: Unmodified 3BNC117 was compared with 3BNC117 bound to the chelator MeCOSar and 64Cu (64Cu-3BNC117) in vitro to assess binding and neutralization. In a clinical trial 64Cu-3BNC117 was infused into HIV uninfected (Group 1), HIV infected and viremic (viral load, VL >1000 c/mL; Group 2) and HIV infected aviremic (VL <20 c/mL; Group 3) participants using two dosing strategies: high protein (3mg/kg unlabeled 3BNC117 combined with <5mg 64Cu-3BNC117) and trace (<5mg 64Cu-3BNC117 only). All participants were screened for 3BNC117 sensitivity from virus obtained from viral outgrowth. Magnetic resonance imaging (MRI)/PET and pharmacokinetic assessments (ELISA for serum 3BNC117 concentrations and gamma counting for 64Cu) were performed 1, 24- and 48-hours post dosing. The trial (clincialtrials.gov NCT03063788) primary endpoint was comparison of PET standard uptake values (SUVs) in regions of interest (e.g lymph node groups and gastrointestinal tract). FINDINGS: Comparison of unmodified and modified 3BNC117 in vitro demonstrated no difference in HIV binding or neutralisation. 17 individuals were enrolled of which 12 were dosed including Group 1 (n=4, 2 high protein, 2 trace dose), Group 2 (n=6, 2 high protein, 4 trace) and Group 3 (n=2, trace only). HIV+ participants had a mean CD4 of 574 cells/microL and mean age 43 years. There were no drug related adverse effects and no differences in tissue uptake in regions of interest (e.g lymph node gut, pharynx) between the 3 groups. In the high protein dosing group, serum concentrations of 3BNC117 and gamma counts were highly correlated demonstrating that 64Cu-3BNC117 remained intact in vivo. INTERPRETATION: In PLWH on or off ART, the intervention of infusing 64Cu-3BNC117 and MRI/PET imaging over 48 hours, was unable to detect HIV-1 env expression in vivo. Future studies should investigate alternative radiolabels such as zirconium which have a longer half-life in vivo. FUNDING: Funded by the Alfred Foundation, The Australian Centre for HIV and Hepatitis Virology Research with additional support from the Division of AIDS, National Institute of Allergy and Infectious Disease, US National Institutes of Health (USAI126611). JHM and SRL are supported by the Australian National Health and Medical Research Council.


Subject(s)
Antibodies, Monoclonal/chemistry , HIV Antibodies/chemistry , HIV Infections/diagnostic imaging , HIV-1/immunology , Radiopharmaceuticals/administration & dosage , Adult , Anti-Retroviral Agents/therapeutic use , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Case-Control Studies , Copper Radioisotopes/chemistry , Female , HIV Antibodies/immunology , HIV Envelope Protein gp120/immunology , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/isolation & purification , HIV-1/metabolism , Half-Life , Humans , Isotope Labeling , Male , Middle Aged , Positron-Emission Tomography , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/immunology , Radiopharmaceuticals/pharmacokinetics , Tomography, X-Ray Computed
4.
Lung Cancer ; 153: 35-41, 2021 03.
Article in English | MEDLINE | ID: mdl-33453471

ABSTRACT

AIM: Conformational forms of the epidermal growth factor receptor (EGFR) are pro-tumorigenic. The prevalence and impact of conformational forms of EGFR in malignant mesothelioma (MM) is unknown. We investigated expression of EGFR and conformational forms of EGFR by immunohistochemistry using EGFR-targeting monoclonal antibodies (mAb). In addition, EGFR gene amplification was investigated by fluorescent in-situ hybridization (FISH). Findings were correlated with survival. METHODS: Patients treated between 1988 and 2014 were identified from the thoracic surgery database of the Austin Hospital, Melbourne, Australia. Tissue microarrays (TMAs) were constructed, subjected to wild type (wt) EGFR IHC staining and FISH analysis. Conformational and mutation forms of EGFR were detected by IHC using mAb806, and LMH-151 which detects EGFRVIII. `H-scores` were derived and EGFR expression correlated with survival by Kaplan-Meier and log rank analysis. RESULTS: WtEGFR expression was seen in 93 % (299/321) of cases with overexpression (defined as an H-score ≥200) seen in more than half of cases (64 %). EGFR overexpression in MM was seen more commonly in the epithelioid subtype. EGFR overexpression was not associated with true EGFR amplification, although multiple copies were appreciated in samples with polysomy. EGFR expression did not correlate with survival. A conformational form of EGFR associated with EGFR dysregulation was found in 8.2 % of cases, and patients with these tumors had a trend towards a poorer outcome. No cases of the EGFRVIII mutation were identified. CONCLUSION: MM consistently demonstrated high expression of EGFR, with a subset of tumors showing conformational EGFR forms consistent with EGFR dysregulation, but withoutEGFR amplification or EGFR VIII mutation. wtEGFR expression did not influence survival. The impact of EGFR conformation on survival warrants further investigation.


Subject(s)
Lung Neoplasms , Mesothelioma, Malignant , Australia , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Amplification , Humans , Lung Neoplasms/genetics , Prognosis
5.
Theranostics ; 10(25): 11404-11415, 2020.
Article in English | MEDLINE | ID: mdl-33052222

ABSTRACT

Through protein engineering and a novel pegylation strategy, a diabody specific to tumor-associated glycoprotein 72 (TAG-72) (PEG-AVP0458) has been created to optimize pharmacokinetics and bioavailability to tumor. We report the preclinical and clinical translation of PEG-AVP0458 to a first-in-human clinical trial of a diabody. Methods: Clinical translation followed characterization of PEG-AVP0458 drug product and preclinical biodistribution and imaging assessments of Iodine-124 trace labeled PEG-AVP0458 (124I-PEG-AVP0458). The primary study objective of the first-in-human study was the safety of a single protein dose of 1.0 or 10 mg/m2 124I-PEG-AVP0458 in patients with TAG-72 positive relapsed/ metastatic prostate or ovarian cancer. Secondary study objectives were evaluation of the biodistribution, tumor uptake, pharmacokinetics and immunogenicity. Patients were infused with a single-dose of 124I labeled PEG-AVP0458 (3-5 mCi (111-185 MBq) for positron emission tomography (PET) imaging, performed sequentially over a one-week period. Safety, pharmacokinetics, biodistribution, and immunogenicity were assessed up to 28 days after infusion. Results: PEG-AVP0458 was radiolabeled with 124I and shown to retain high TAG-72 affinity and excellent targeting of TAG-72 positive xenografts by biodistribution analysis and PET imaging. In the first-in-human trial, no adverse events or toxicity attributable to 124I-PEG-AVP0458 were observed. Imaging was evaluable in 5 patients, with rapid and highly specific targeting of tumor and minimal normal organ uptake, leading to high tumor:blood ratios. Serum concentration values of 124I-PEG-AVP0458 showed consistent values between patients, and there was no significant difference in T½α and T½ß between dose levels with mean (± SD) results of T½α = 5.10 ± 4.58 hours, T½ß = 46.19 ± 13.06 hours. Conclusions: These data demonstrates the safety and feasibility of using pegylated diabodies for selective tumor imaging and potential delivery of therapeutic payloads in cancer patients.


Subject(s)
Antibodies, Bispecific/adverse effects , Antigens, Neoplasm/metabolism , Antineoplastic Agents/adverse effects , Neoplasms/drug therapy , Radiopharmaceuticals/adverse effects , Adult , Animals , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/genetics , Antibodies, Bispecific/pharmacokinetics , Antibodies, Neoplasm/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Cell Line, Tumor , Dose-Response Relationship, Drug , Feasibility Studies , Female , Humans , Infusions, Intravenous , Iodine Radioisotopes/administration & dosage , Iodine Radioisotopes/adverse effects , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Neoplasms/diagnosis , Neoplasms/immunology , Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Single-Chain Antibodies/administration & dosage , Single-Chain Antibodies/adverse effects , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacokinetics , Tissue Distribution , Xenograft Model Antitumor Assays
6.
PLoS One ; 13(7): e0201089, 2018.
Article in English | MEDLINE | ID: mdl-30052660

ABSTRACT

PDGF-CC is a member of the platelet-derived growth factor (PDGF) family that stimulates PDGFRα phosphorylation and thereby activates intracellular signalling events essential for development but also in cancer, fibrosis and neuropathologies involving blood-brain barrier (BBB) disruption. In order to elucidate the biological and pathological role(s) of PDGF-CC signalling, we have generated high affinity neutralizing monoclonal antibodies (mAbs) recognizing human PDGF-CC. We determined the complementarity determining regions (CDRs) of the selected clones, and mapped the binding epitope for clone 6B3. Using the monoclonal 6B3, we determined the expression pattern for PDGF-CC in different human primary tumours and control tissues, and explored its ability to neutralize PDGF-CC-induced phosphorylation of PDGFRα. In addition, we showed that PDGF-CC induced disruption of the blood-retinal barrier (BRB) was significantly reduced upon intraperitoneal administration of a chimeric anti-PDGF-CC antibody. In summary, we report on high affinity monoclonal antibodies against PDGF-CC that have therapeutic efficacy in vivo.


Subject(s)
Antibodies, Monoclonal/immunology , Lymphokines/antagonists & inhibitors , Lymphokines/immunology , Platelet-Derived Growth Factor/antagonists & inhibitors , Platelet-Derived Growth Factor/immunology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , A549 Cells , Animals , Blood-Retinal Barrier/drug effects , Blood-Retinal Barrier/metabolism , Blood-Retinal Barrier/pathology , Capillary Permeability , Gene Expression/drug effects , Humans , Immunohistochemistry , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Proteins/immunology , Signal Transduction/drug effects
7.
Apoptosis ; 20(11): 1483-98, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26342814

ABSTRACT

Fibroblast activation protein (FAP), an integral membrane serine protease, is found on fibro- and osteo-sarcoma and on myofibroblasts in epithelial carcinoma, but rarely on other adult tissue. FAP has been demonstrated to be an excellent target for tumor imaging in clinical trials, and antibodies and other FAP-targeting drugs are in development. Here we have shown that FAP overexpression increased the growth of HT1080 fibrosarcoma cells in vitro and in vivo, and found that the expression of FAP affects response to chemotherapy. When treated with doxorubicin, expression of FAP increased susceptibility to the drug. In spite of this, FAP-HT1080 cells had fewer markers of classical apoptosis than HT1080 cells and neither necrosis nor necroptosis were enhanced. However, levels of early mitochondrial and lysosomal membrane permeability markers were increased, and autophagy switched from a protective function in HT1080 cells to part of the cell death mechanism with FAP expression. Therefore, FAP may affect how the tumor responds to chemotherapeutic drugs overall, which should be considered in targeted drug development. The overexpression of FAP also alters cell signaling and responses to the environment in this cell line. This includes cell death mechanisms, changing the response of HT1080 cells to doxorubicin from classical apoptosis to an organelle membrane permeability-dependent form of cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fibrosarcoma/enzymology , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Autophagy/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Endopeptidases , Fibrosarcoma/drug therapy , Fibrosarcoma/genetics , Fibrosarcoma/physiopathology , Gelatinases/genetics , Humans , Membrane Proteins/genetics , Serine Endopeptidases/genetics
8.
J Clin Oncol ; 33(24): 2609-16, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26124477

ABSTRACT

PURPOSE: CS-1008 (tigatuzumab) is a humanized, monoclonal immunoglobulin G1 (IgG1) agonistic antibody to human death receptor 5. The purpose of this study was to investigate the impact of CS-1008 dose on the biodistribution, quantitative tumor uptake, and antitumor response in patients with metastatic colorectal cancer (mCRC). PATIENTS AND METHODS: Patients with mCRC who had received at least one course of chemotherapy were assigned to one of five dosage cohorts and infused with a weekly dose of CS-1008. Day 1 and day 36 doses were trace-labeled with indium-111 ((111)In), followed by whole-body planar and regional single-photon emission computed tomography (SPECT) imaging at several time points over the course of 10 days. RESULTS: Nineteen patients were enrolled. (111)In-CS-1008 uptake in tumor was observed in only 12 patients (63%). (111)In-CS-1008 uptake and pharmacokinetics were not affected by dose or repeated drug administration. (111)In-CS-1008 biodistribution showed gradual blood-pool clearance and no abnormal uptake in normal tissue. No anti-CS-1008 antibody development was detected. One patient achieved partial response (3.7 months duration), eight patients had stable disease, and 10 patients had progressive disease. Clinical benefit rate (stable disease + partial response) in patients with (111)In-CS-1008 uptake in tumor was 58% versus 28% in patients with no uptake. An analysis of individual lesions showed that lesions with antibody uptake were one third as likely to progress as those without antibody uptake (P = .07). Death-receptor-5 expression in archived tumor samples did not correlate with (111)In-CS-1008 uptake (P = .5) or tumor response (P = .6). CONCLUSION: Death-receptor-5 imaging with (111)In-CS-1008 reveals interpatient and intrapatient heterogeneity of uptake in tumor, is not dose dependent, and is predictive of clinical benefit in the treatment of patients who have mCRC.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/drug therapy , Aged , Aged, 80 and over , Cohort Studies , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , Humans , Indium Radioisotopes , Male , Middle Aged , Neoplasm Metastasis , Radionuclide Imaging , Radiopharmaceuticals/administration & dosage , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution
9.
Clin Exp Metastasis ; 32(5): 507-16, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25995078

ABSTRACT

The serine protease fibroblast activation protein (FAP) is selectively expressed on tumour-associated fibroblasts in most human epithelial tumours, as well as on some mesenchymal tumours such as sarcoma. High FAP expression is most often associated with poor outcome and increased metastasis. Here, we compare the in vitro metastatic potential of HT1080 fibrosarcoma cells with and without FAP expression in order to elucidate the mechanism by which FAP may influence metastasis. In the presence of FAP, cells were more adhesive to extracellular matrix proteins and migrated and invaded through Matrigel to a greater degree. The anti-FAP antibody ESC11, which caused internalization of FAP, decreased adhesion and migration, but only when cells expressed FAP. It was also found that blocking activity of integrins ß1 and αvß3 reduced both cell adhesion and migration and this effect was much more marked in FAP-expressing HT1080 cells than mock-transfected HT1080 cells. The expression or activation of intracellular proteins that form part of the downstream signaling of integrins, including integrin-linked kinase, Rac1 and focal adhesion kinase, was also upregulated when FAP was expressed, suggesting that FAP not only upregulates metastatic-like cell behaviours through interaction with integrins, but also influences the intracellular signaling of integrins. This was confirmed using both PI3 kinase and Src kinase inhibitors, which decreased adhesion and migration in FAP-expressing cells, but did not affect mock-transfected HT1080 cells. FAP is therefore a useful target for anti-cancer therapy, as not only is its expression tumour-selective, but its downregulation has the potential to reduce incidence of metastasis.


Subject(s)
Cell Movement , Cell Proliferation , Fibrosarcoma/metabolism , Fibrosarcoma/secondary , Gelatinases/metabolism , Integrin alphaVbeta3/metabolism , Integrin beta1/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cell Adhesion , Endopeptidases , Flow Cytometry , Gelatinases/antagonists & inhibitors , Gelatinases/immunology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Microscopy, Fluorescence , Neoplasm Invasiveness , Serine Endopeptidases/immunology , Tumor Cells, Cultured
10.
J Nucl Med ; 55(4): 534-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24556590

ABSTRACT

UNLABELLED: huA33 is a humanized antibody that targets the A33 antigen, which is highly expressed in intestinal epithelium and more than 95% of human colon cancers but not other normal tissues. Previous studies have shown huA33 can target and be retained in a metastatic tumor for 6 wk but eliminated from normal colonocytes within days. This phase I study used radiolabeled huA33 in combination with capecitabine to target chemoradiation to metastatic colorectal cancer. The primary objective was safety and tolerability of the combination of capecitabine and (131)I-huA33. Pharmacokinetics, biodistribution, immunogenicity, and tumor response were also assessed. METHODS: Eligibility included measurable metastatic colorectal cancer, adequate hematologic and biochemical function, and informed consent. An outpatient scout (131)I-huA33 dose was followed by a single-therapy infusion 1 wk later, when capecitabine was commenced. Dose escalation occurred over 5 dose levels. Patients were evaluated weekly, with tumor response assessment at the end of the 12-wk trial. Tumor targeting was assessed using a γ camera and SPECT imaging. RESULTS: Nineteen eligible patients were enrolled. The most frequently observed toxicity included myelosuppression, gastrointestinal symptoms, and asymptomatic hyperbilirubinemia. Biodistribution analysis demonstrated excellent tumor targeting of the known tumor sites, expected transient bowel uptake, but no other normal tissue uptake. (131)I-huA33 demonstrated a mean terminal half-life and serum clearance suited to radioimmunotherapy (T1/2ß, 100.24 ± 20.92 h, and clearance, 36.72 ± 8.01 mL/h). The mean total tumor dose was 13.8 ± 7.6 Gy (range, 5.1-26.9 Gy). One patient had a partial response, and 10 patients had stable disease. CONCLUSION: (131)I-huA33 achieves specific targeting of radiotherapy to colorectal cancer metastases and can be safely combined with chemotherapy, providing an opportunity to deliver chemoradiation specifically to metastatic disease in colorectal cancer patients.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Chemoradiotherapy/methods , Colorectal Neoplasms/radiotherapy , Colorectal Neoplasms/therapy , Radiopharmaceuticals/therapeutic use , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Neoplasm/immunology , Antimetabolites, Antineoplastic/therapeutic use , Capecitabine , Colorectal Neoplasms/immunology , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Female , Fluorouracil/analogs & derivatives , Fluorouracil/therapeutic use , Humans , Isotope Labeling , Male , Middle Aged , Neoplasm Metastasis , Patient Selection , Radiometry , Radiopharmaceuticals/adverse effects , Radiopharmaceuticals/pharmacokinetics , Tissue Distribution , Tomography, X-Ray Computed
11.
Neuropsychologia ; 49(9): 2685-93, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21651922

ABSTRACT

Individuals born very preterm (VPT) are at risk of neurodevelopmental damage and of adverse educational outcomes in childhood and adolescence. The present study used voxel-based morphometry to investigate the association between grey matter and white matter volume and measures of language and executive functioning in VPT born adolescents and term-born controls by gender. VPT individuals (N=218) and controls (N=127) underwent neuropsychological assessment and MRI at age 14-15 as part of a longitudinal study. Differential associations were found between spelling scores and frontal regional grey matter volume when group (VPT and control) and gender (males and females) were investigated. A main effect of group demonstrated a weaker association in VPT adolescents relative to controls between grey matter volume in the left medial and right superior frontal gyri and spelling scores. A main effect of gender revealed spelling scores to be correlated with grey matter volume in the right superior frontal gyrus in females to a greater extent than in males. Furthermore, a significant interaction between group and gender was detected in two regions. Spelling scores showed a stronger association with grey matter volume in a cluster with local maxima in the left medial frontal cortex extending to the caudate nucleus in VPT females than in control females and a weaker association in VPT males compared to control males. In addition, spelling scores showed a stronger association with grey matter volume in left middle frontal gyrus in VPT males compared to control males and a weaker association in VPT females than in control females. When group and gender were investigated, there were no statistically different correlations between structural brain volumes and performance on reading and executive function tests. These data demonstrate that the typical structure-function relationship in respect to spelling abilities appears to be altered in individuals born preterm and the processes underpinning this divergence may be subject to gender-specific influences.


Subject(s)
Brain/anatomy & histology , Cognition Disorders/pathology , Frontal Lobe/anatomy & histology , Language Disorders/pathology , Adolescent , Brain/physiology , Case-Control Studies , Cognition Disorders/physiopathology , Cohort Studies , Female , Frontal Lobe/physiology , Humans , Infant, Newborn , Language Disorders/physiopathology , Longitudinal Studies , Male , Neuropsychological Tests , Organ Size , Premature Birth , Reference Values , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL