Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Cancer Med ; 12(11): 12095-12105, 2023 06.
Article in English | MEDLINE | ID: mdl-37012214

ABSTRACT

BACKGROUND: Since it was first reported in December 2019, coronavirus disease 2019 (COVID-19) spread rapidly across the globe resulting in a pandemic. As of August 2022, seven outbreak peaks have been confirmed in Tokyo, and the numbers of new cases in the fifth and later outbreak periods have been far greater than in the preceding periods. This retrospective study examined the impact of the COVID-19 pandemic on perioperative chemotherapy for breast cancer. METHODS: Patients with breast cancer who received perioperative chemotherapy at the National Cancer Center Hospital East were divided into 2 groups: 120 and 384 patients who started chemotherapy before and during the pandemic, respectively. The incidence of critical events that had potential detrimental effects on the prognosis, such as start of adjuvant chemotherapy ≥91 days after surgery and relative dose intensity of chemotherapy <85% were compared between groups. RESULTS: No significant difference in the incidence of critical events was found. When stratified by outbreak period, the incidence of critical events was positively correlated with the increasing number of new cases of COVID-19 (r = 0.83, p = 0.04). Moreover, 25/173 patients (14%) who started perioperative chemotherapy during the fifth and sixth outbreak periods developed COVID-19 infection, 80% of whom (20/25) had a delay or interruption to their surgery or other perioperative treatments. CONCLUSIONS: Although the impact of the COVID-19 pandemic on perioperative chemotherapy on whole groups of patients was not evident when comparing periods before and after the pandemic, the impact is becoming prominent in parallel with increasing numbers of new COVID-19 cases.


Subject(s)
Breast Neoplasms , COVID-19 , Humans , Female , COVID-19/epidemiology , Pandemics , SARS-CoV-2 , Breast Neoplasms/drug therapy , Breast Neoplasms/epidemiology , Breast Neoplasms/surgery , Retrospective Studies
2.
Jpn J Clin Oncol ; 53(2): 105-114, 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36373891

ABSTRACT

BACKGROUND: Cyclin-dependent kinase 4/6 (CDK4/6) inhibitors are the standard treatment for advanced hormone receptor-positive breast cancer. Although interstitial lung disease is a rare (1-3.3%) but serious adverse event associated with CDK4/6 inhibitors, the incidence of interstitial lung disease in Japanese patients in the real world and the risk factors of interstitial lung disease are not clear. METHODS: We retrospectively investigated the incidence of interstitial lung disease in 224 patients with advanced breast cancer who received CDK4/6 inhibitors at our hospital between 31 January 2017 and 31 January 2021. The correlation of age (>50 vs ≤50 years), presence or absence of previous history of interstitial lung disease, lung metastasis, smoking history and chest radiation with the development of interstitial lung disease was evaluated. RESULTS: In total, 177 cases received palbociclib, 39 cases received abemaciclib and 8 cases received both palbociclib and abemaciclib, constituting a palbociclib group (n = 185) and an abemaciclib group (n = 47). At a median observation period of 607 days, 8.0% (18/224) cases (13 definite and 5 probable cases) had interstitial lung disease; 6.5% (12/185) of palbociclib-treated and 13% (6/47) of abemaciclib-treated cases. The median time to interstitial lung disease onset was 178 (range, 14-750) days. There was no significant correlation between the background factors studied and the development of interstitial lung disease. CONCLUSION: The frequency of CDK4/6 inhibitor-induced interstitial lung disease was higher than that reported in clinical trials. We did not identify any risk factors for the development of interstitial lung disease in this study, and thus, larger studies that include patient predisposition are required.


Subject(s)
Breast Neoplasms , Protein Kinase Inhibitors , Female , Humans , Middle Aged , Aminopyridines/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Cyclin-Dependent Kinase 6/antagonists & inhibitors
3.
Infez Med ; 25(2): 169-173, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28603238

ABSTRACT

We describe the case of a 67-year-old male with liver cirrhosis who presented with fever and neck pain. Magnetic resonance imaging of the spine detected cervical vertebral osteomyelitis, and enhanced CT of the neck and spine revealed retropharyngeal abscess. The patient was treated with empirical antimicrobial therapy and surgical drainage due to significant airway involvement. Escherichia coli was cultured from the blood and pus in inferior cervical vertebrae which was a rare pathogen. Haematogenous spread may have resulted in cervical vertebral osteomyelitis and retropharyngeal abscess. With high mortality rates, early diagnosis of retropharyngeal abscess is required to avoid debilitating complications such as airway obstruction.


Subject(s)
Cervical Vertebrae/microbiology , Discitis/etiology , Escherichia coli Infections/etiology , Liver Cirrhosis, Alcoholic/complications , Retropharyngeal Abscess/etiology , Aged , Airway Obstruction/etiology , Anti-Bacterial Agents/therapeutic use , Bacteremia/drug therapy , Bacteremia/etiology , Cephalosporins/therapeutic use , Discitis/microbiology , Discitis/surgery , Disease Susceptibility , Drainage , Drug Substitution , Escherichia coli Infections/drug therapy , Escherichia coli Infections/surgery , Humans , Magnetic Resonance Imaging , Male , Neck Pain/etiology , Osteomyelitis/drug therapy , Osteomyelitis/microbiology , Osteomyelitis/surgery , Oxygen Inhalation Therapy , Retropharyngeal Abscess/diagnostic imaging , Retropharyngeal Abscess/microbiology , Retropharyngeal Abscess/surgery , Tomography, X-Ray Computed
4.
Infez Med ; 23(2): 168-73, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26110298

ABSTRACT

We report a case of renal embolism as an initial manifestation of Streptococcus dysgalactiae subspecies equisimilis (SDSE) endocarditis in a patient with chronic aortic dissection. A 37-year-old man who underwent total aortic arch replacement owing to aortic dissection, presented with a 3-h history of fever, chills, and acute right-sided flank pain. The endocarditis affected the native aortic valve and was complicated by a renal embolism. Blood culture results were positive for SDSE. Intravenous penicillin resulted in satisfactory clinical and echocardiographic recovery.


Subject(s)
Aortic Aneurysm/complications , Endocarditis/complications , Renal Artery Obstruction/microbiology , Streptococcal Infections/complications , Streptococcus/isolation & purification , Thromboembolism/microbiology , Administration, Intravenous , Adult , Aortic Dissection/complications , Anti-Bacterial Agents/administration & dosage , Chronic Disease , Endocarditis/drug therapy , Endocarditis/microbiology , Humans , Male , Penicillins/administration & dosage , Renal Artery Obstruction/complications , Streptococcal Infections/drug therapy , Streptococcus/classification , Streptococcus/pathogenicity , Thromboembolism/complications , Treatment Outcome
5.
Mol Cancer Ther ; 12(4): 384-93, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358664

ABSTRACT

HER2 is a transmembrane oncoprotein encoded by the HER2/neu gene and is overexpressed in approximately 20% to 30% of breast cancers. We have recently designed a novel class of drug, the hybrid peptide, which is chemically synthesized and is composed of a target-binding peptide and a lytic peptide containing cationic-rich amino acid components that disintegrate the cell membrane, leading to cancer cell death via membrane lysis. In this study, we designed a HER2-binding peptide linked to this novel lytic peptide, which we termed the HER2-lytic hybrid peptide and assessed the cytotoxic activity of this hybrid peptide in vitro and in vivo. The HER2-lytic hybrid peptide showed high cytotoxic activity against all ovarian and breast cancer cell lines, even trastuzumab- and/or lapatinib-resistant cells, but not against normal cells. Competition assays using anti-HER2 antibody and knockdown of this receptor by siRNA confirmed the specificity of the HER2-lytic hybrid peptide. In addition, it was shown that the HER2-lytic hybrid peptide can disintegrate the cancer cell membrane of HER2-overexpressing SK-BR-3 cancer cells in only 5 minutes, but not normal cells, and block HER2 signaling. Intravenous administration of the HER2-lytic peptide in the athymic mouse implanted with BT-474 and MDA-MB-453 cells significantly inhibited tumor progression. The HER2-lytic hybrid peptide was effective even in breast cancer cell lines that are resistant to trastuzumab and/or lapatinib in vitro and in vivo. Therefore, this hybrid peptide may provide a potent treatment option for patients with cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Membrane/metabolism , Peptides/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Humans , Mice , Peptides/metabolism , Peptides/toxicity , Protein Binding , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Chemother Pharmacol ; 71(3): 799-807, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23328867

ABSTRACT

PURPOSE: Transferrin receptor (TfR) is a cell membrane-associated glycoprotein involved in the cellular uptake of iron and the regulation of cell growth. Recent studies have shown elevated expression levels of TfR on cancer cells compared with normal cells. We previously designed a TfR-lytic hybrid peptide, which combines the TfR-binding peptide and a lytic peptide, and reported that it bound specifically to TfR and selectively killed cancer cells. Furthermore, the intravenous administration of TfR-lytic peptide in an athymic mouse model significantly inhibited tumor progression. To evaluate the immunogenicity of this peptide as a novel and potent anticancer agent, we investigated whether TfR-lytic hybrid peptide elicits cellular and humoral immune responses to produce antibodies. We also examined the toxicity of this peptide in syngeneic mice. METHODS: We performed hematologic and blood chemistry test and histological analysis and assessed hemolytic activity to check toxicity. To evaluate the immunogenicity, measurement of murine interferon-gamma and detection of TfR-lytic-specific antibody by ELISA were demonstrated. RESULTS: No T cell immune response or antibodies were detected in the group treated with TfR-lytic hybrid peptide. No hematologic toxicity, except for a decrease in leukocytes, was observed, and no remarkable influence on metabolic parameters and organs (liver, kidney, and spleen) was noted. CONCLUSIONS: Therefore, TfR-lytic hybrid peptide might provide an alternative therapeutic option for patients with cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides/pharmacology , Receptors, Transferrin/drug effects , Amino Acid Sequence , Animals , Antineoplastic Agents/immunology , Cell Line, Tumor , Erythrocytes/drug effects , Female , Flow Cytometry , Hemolysis/drug effects , Humans , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Interferon-gamma/pharmacology , Lymph Nodes/cytology , Lymph Nodes/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasm Transplantation , Peptides/immunology , Transplantation, Isogeneic
7.
J Biosci Bioeng ; 114(1): 96-103, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22425524

ABSTRACT

We previously reported that Antp-TPR hybrid peptide inhibited the interaction of Hsp90 with TPR2A and had selective cytotoxic activity discriminating between normal and cancer cells to induce cancer cell death. In this study, we investigated the cytotoxic activity of Antp-TPR peptide toward acute myeloid leukemia (AML) cells. It was demonstrated that Antp-TPR peptide induced AML cell death in cell lines such as U937, K562, THP-1, and HL-60 via activation of caspases 3 and 7, and disruption of mitochondrial membrane potential. Conversely, Antp-TPR peptide did not reduce the viability of normal cells including peripheral blood mononuclear cells (PBMCs), although both geldanamycin and 17-AAG, small-molecule inhibitors of Hsp90, mediated cytotoxicity to these normal cells at low concentrations. In addition, mutation analysis of TPR peptide demonstrated that the highly conserved amino acids Lys and Arg were critical to the cytotoxic activity. These results indicated that Antp-TPR hybrid peptide would provide potent and selective therapeutic options in the treatment of AML.


Subject(s)
Cytotoxins/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Homeodomain Proteins/pharmacology , Leukocytes, Mononuclear/drug effects , Amino Acid Sequence , Amino Acids/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , HEK293 Cells , HL-60 Cells , HSP90 Heat-Shock Proteins/chemistry , Homeodomain Proteins/chemistry , Homeodomain Proteins/metabolism , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Inhibitory Concentration 50 , K562 Cells , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/cytology , MCF-7 Cells , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment , Survivin , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , U937 Cells
8.
BMC Cancer ; 11: 359, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21849092

ABSTRACT

BACKGROUND: Transferrin receptor (TfR) is a cell membrane-associated glycoprotein involved in the cellular uptake of iron and the regulation of cell growth. Recent studies have shown the elevated expression levels of TfR on cancer cells compared with normal cells. The elevated expression levels of this receptor in malignancies, which is the accessible extracellular protein, can be a fascinating target for the treatment of cancer. We have recently designed novel type of immunotoxin, termed "hybrid peptide", which is chemically synthesized and is composed of target-binding peptide and lytic peptide containing cationic-rich amino acids components that disintegrates the cell membrane for the cancer cell killing. The lytic peptide is newly designed to induce rapid killing of cancer cells due to conformational change. In this study, we designed TfR binding peptide connected with this novel lytic peptide and assessed the cytotoxic activity in vitro and in vivo. METHODS: In vitro: We assessed the cytotoxicity of TfR-lytic hybrid peptide for 12 cancer and 2 normal cell lines. The specificity for TfR is demonstrated by competitive assay using TfR antibody and siRNA. In addition, we performed analysis of confocal fluorescence microscopy and apoptosis assay by Annexin-V binding, caspase activity, and JC-1 staining to assess the change in mitochondria membrane potential. In vivo: TfR-lytic was administered intravenously in an athymic mice model with MDA-MB-231 cells. After three weeks tumor sections were histologically analyzed. RESULTS: The TfR-lytic hybrid peptide showed cytotoxic activity in 12 cancer cell lines, with IC(50) values as low as 4.0-9.3 µM. Normal cells were less sensitive to this molecule, with IC(50) values > 50 µM. Competition assay using TfR antibody and knockdown of this receptor by siRNA confirmed the specificity of the TfR-lytic hybrid peptide. In addition, it was revealed that this molecule can disintegrate the cell membrane of T47D cancer cells just in 10 min, to effectively kill these cells and induce approximately 80% apoptotic cell death but not in normal cells. The intravenous administration of TfR-lytic peptide in the athymic mice model significantly inhibited tumor progression. CONCLUSIONS: TfR-lytic peptide might provide a potent and selective anticancer therapy for patients.


Subject(s)
Cell Membrane/drug effects , Immunotoxins/pharmacology , Neoplasms/drug therapy , Peptides/pharmacology , Receptors, Transferrin/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Female , Humans , Immunotoxins/metabolism , Inhibitory Concentration 50 , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Molecular Sequence Data , Neoplasms/metabolism , Neoplasms/pathology , Peptides/genetics , Peptides/metabolism , Protein Binding , RNA Interference , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Spleen/drug effects , Spleen/metabolism , Spleen/pathology , Time Factors , Xenograft Model Antitumor Assays
9.
Oncol Rep ; 25(2): 469-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21165574

ABSTRACT

Cancer immunotherapy is a potential therapeutic strategy, in addition to surgical treatment, radiotherapy, and chemotherapy. Cancer-specific immunotherapy, such as the MAGE peptide vaccine, has been utilized clinically. How-ever, there are inherent limits to the effectiveness of vaccinotherapy using a single antigen because of the expression frequency of cancer-specific antigens on tumor cells. Thus, identification of a new cancer-specific antigen is needed. In this study, we examined the possibility of using cancer-specific immunotherapy based upon mitotic centromere-associated kinesin (MCAK) which was previously identified as a novel cancer/testis antigen. To evaluate the feasibility of developing cancer immunotherapy using MCAK peptides, we studied HLA-A*0201 and *2402 as targets for CTLs in the context of HLA class I molecules. By using a peptide with a sequence of AINPELLQL (amino acid positions 63-71 in MCAK, HLA-A*0201) and FFEIYNGKL (amino acid positions 401-409 in MCAK, HLA-A*2402), CTL responses could be induced from unseparated PBMCs by stimulation of freshly isolated, peptide-pulsed PBMCs as antigen-presenting cells (APCs) and also by using interleukin-7 and keyhole limpet hemocyanin in primary culture. The induced CTLs could lyse HLA-A-*0201/*2402 colon and gastric cancer cells expressing MCAK, as well as the peptide-pulsed target cells, in an HLA class l, and CD8 restricted manner. The identification of the MCAK/HLA-A*0201 and *2402 peptides suggests the possibility of designing peptide-based immunotherapeutic approaches that might prove effective in treating patients with MCAK-positive cancer.


Subject(s)
Epitopes, T-Lymphocyte/isolation & purification , Immunity, Cellular/immunology , Kinesins/chemistry , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cell Culture Techniques , Epitope Mapping , Epitopes, T-Lymphocyte/immunology , HLA-A Antigens/metabolism , HLA-A2 Antigen , HLA-A24 Antigen , Humans , Immunity, Cellular/genetics , K562 Cells , Kinesins/genetics , Kinesins/immunology , Kinesins/metabolism , Neoplasms/genetics , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptide Fragments/isolation & purification , Substrate Specificity/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...