Kentaro Sudo’s research while affiliated with Chiba Cancer Center and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (44)


Diagnosis of pancreatic acinar cell carcinoma (ACC). (A) Diagnostic images of an ACC case. (Left) Enhanced computed tomography (CT). The tumor is a low‐density mass in the pancreatic head (arrowheads). (Middle) Endoscopic retrograde cholangiopancreatography (ERCP). The bile duct stenosis (arrowheads) is caused by the tumor. (Right) Endoscopic ultrasound‐guided fine‐needle aspiration (EUS‐FNA). The tumor appears to be hypoechoic (arrowheads). A biopsy specimen is aspirated with the needle (arrow). (B) Histological analysis of the biopsy specimen. (Left) Hematoxylin and eosin (H&E) staining. (Middle) Immunostaining of carboxyl ester hydrolase (CEH) with anti‐BCL10 mAb. (Right) Immunostaining for synaptophysin. Scale bars, 50 μm. (C) The resected tumor. An encapsulated mass (arrowheads) is in the middle of the resected tissue. (D) Histological analysis of the resected tumor. (Left) H&E staining. (Right) Immunostaining of CEH with anti‐BCL10 mAb. Scale bars, 100 μm.
Propagation of patient‐derived organoids (PDOs) and development of a xenograft. (A) Schematic view of a Matrigel bilayer organoid culture (MBOC). (B) Phase contrast images of PDOs from different sources. These PDOs are designated as bile‐, biopsy‐, and surgery‐PDO, respectively. Scale bars, 200 μm. (C) Xenograft development. The tumor originates from the biopsy‐PDO. Scale bar, 5 mm. (D) Histological analysis of the xenograft. (Left) H&E staining. (Right) Immunostaining of CEH with anti‐BCL10 mAb. Scale bar, 50 μm. (E) Phase contrast images of the xenograft‐derived organoid (XDO). (F) Histological analysis of the XDO. (Left) H&E staining. (Right) immunostaining of CEH with anti‐BCL10 mAb. Insets show a magnified image of representative organoids. Scale bars, 50 μm. (G) Western blotting of organoids. Note that anti‐BCL10 mAb (clone 331.3) detects both BCL10 (33 kDa) and CEH (105 kDa). α‐Tubulin serves as a loading control. NS, non‐specific bands. An image by long exposure is also shown (left). (H) Schematic summary of establishment of PDOs and the XDO (HS‐1). Color intensity of the arrows reflects proliferation rate. NT, not tested for inoculation in nude mice.
Propagation of HS‐1 from the biopsy‐PDO. (A) Effect of culture conditions. Phase contrast images of HS‐1 cultured under various conditions. Organoid culture medium is supplemented with 10% FBS or defined growth factors (DGF) without serum. Scale bars, 250 μm. (B) Cell proliferation of HS‐1 in culture. The cell numbers were counted using the trypan blue dye exclusion method. (C) Phase contrast images of HS‐1 in the medium with DGF in the absence of Matrigel. Small aggregates are distributed on the surface of the well, while large aggregates expand upward (arrow). Scale bars, 50 μm. (D) Dual staining of cell aggregates. Semi‐floating and floating cells are stained. Dead cells (red) and viable cells (green) are differentially stained. (E) Schematic illustration of the HS‐1 propagation. Cells grew in a spherical shape in Matrigel, whereas the aggregate grew larger and eventually detached from the dish (arrow) in the absence of Matrigel. (F) Western blotting of the lysate and supernatant of HS‐1. Ponceau S staining of the membrane serves as a loading control. (G) Western blotting of the culture supernatant. Time‐lapse samples are analyzed for secreted trypsin.
Genomic analysis of the ACC tumor and derived organoids. (A) Mutations in the resected ACC tumor and derived organoids. Genes with a variant allele frequency (VAF) >10% estimated using next‐generation sequencing (NGS) analysis are shown. Loss of a 9‐bp deletion in CDKN2A (asterisk) in XDO (HS‐1) was due to deletion of both alleles (see D and E). (B) Validation of somatic mutations in surgery‐PDO. Sanger sequencing verified a homozygous point mutation in EP400 (arrowhead) and a hemizygous 9‐bp deletion in CDKN2A (arrow). (C) Genome‐wide distribution of loss of heterozygosity (LOH). LOH is estimated based on the change in VAF and collectively shown as the line. Gray bars indicate areas with no corresponding data in NGS. (D) Genome‐wide copy number variation (CNV) by array CGH analysis. The regions in which the XDO (HS‐1) acquired copy number gain and loss compared with surgery‐PDO are indicated by (*) and (#), respectively. The 33‐Mb region in the chr. 9p is homozygously deleted in the XDO (HS‐1). Note that chr. 9p in surgery‐PDO seems to undergo slight copy number loss (arrowhead). (E) Genomic PCR of the chr. 9p region. CDKN2A and DNAJA1 are amplified, from inside and outside of the deleted region, respectively. Note that CDKN2A amplicon is faintly visible for XDO (HS‐1). (F) Schematic summary of the genome analysis of tumor and organoids.
Drug sensitivity assay with HS‐1 in vitro and in vivo. (A) Schematic presentation of the drug screening. Organoids grown on Matrigel were treated. (B) Sensitivity of HS‐1 to conventional chemotherapeutic drugs. Error bars depict SD (n = 3). (C) Imaging‐based evaluation of cell viability. Viable cells (green) and dead cells (red) after pneumothorax (PTX) treatment are computationally detected. Scale bar, 500 μm. (D) Correlation between the area of viable organoids and luminescence intensity. Data obtained in the ATP‐based viability assay in (B) were compared with the area of viable cells estimated by computational detection. (E) Imaging‐based drug screening. In total, 364 reagents were administered at concentration of 10 nM. The bars corresponding to bafilomycin and bortezomib are indicated by arrows. (F) Imaging‐based validation of hits in the drug screening. Error bars depict SD (n = 3). (G) Antitumorigenic effect of bortezomib in vivo. After inoculation of HS‐1 to nude mice, 5 μg of bortezomib in saline was administered twice a week (arrows). Error bars depict SD (n = 3 each). *p < 0.05.

+1

Derivation of Pancreatic Acinar Cell Carcinoma Cell Line HS ‐1 as a Patient‐derived Tumor Organoid
  • Article
  • Full-text available

November 2022

·

57 Reads

·

5 Citations

·

·

Yoshiaki Maru

·

[...]

·

Acinar cell carcinoma (ACC) of the pancreas is a malignant tumor of the exocrine cell lineage with a poor prognosis. Due to its rare incidence and technical difficulties, few authentic human cell lines are currently available, hampering detailed investigations of ACC. Therefore, we applied the organoid culture technique to various types of specimens obtained from a single ACC patient, such as bile, biopsy, and resected tumor. Despite initial propagation, none of these organoids achieved long-term proliferation or tolerated cryopreservation, confirming the challenging nature of establishing ACC cell lines. Nevertheless, the biopsy-derived early passage organoid developed subcutaneous tumors in immunodeficient mice. The xenograft tumor histologically resembled the original tumor and gave rise to infinitely propagating organoids with solid features and high levels of trypsin secretion. Moreover, the organoid stained positive for carboxylic ester hydrolase, a specific ACC marker, but negative for the duct cell marker CD133 and the endocrine lineage marker synaptophysin. Hence, we concluded the derivation of a novel ACC cell line of pure exocrine lineage, designated as HS-1. Genomic analysis revealed extensive copy number alterations and mutations in EP400 in the original tumor, which were enriched in primary organoids. HS-1 displayed homozygous deletion of CDKN2A, which might underlie xenograft formation from organoids. Although resistant to standard cytotoxic agents, the cell line was highly sensitive to the proteasome inhibitor bortezomib, as revealed by an in vitro drug screen and in vivo validation. Summarily, we document a novel ACC cell line, which could be useful for ACC studies in the future.

Download

Endoscopic ultrasound-guided tissue acquisition and gene panel testing for pancreatic cancerEUS-guided tissue acquisitionに基づく膵癌遺伝子パネル検査の実際

February 2022

·

11 Reads

Suizo

In the era of genomic medicine, there is a growing need to obtain tissue samples suitable for tumor genotyping. Recent studies have focused on the development of optimal devices and techniques in endoscopic ultrasound-guided tissue acquisition (EUS-TA) for pancreatic cancer. In addition to sufficient tissue acquisition, it is also necessary to optimize preanalytical steps (e.g. sample processing, formalin fixation, and specimen preparation for analysis) to improve the success rate of gene panel testing. Close cooperation with pathologists and technicians is essential for successful analysis. However, in some cases, it is still challenging to obtain sufficient tissue samples. In such cases, we should consider other methods of genomic testing such as plasma cell-free DNA genotyping, microsatellite instability testing, and germline BRCA1/2 testing depending on the patient's condition. In this review, we discuss the role of EUS-TA in the clinical practice of gene panel testing for pancreatic cancer.


Figure 1. Consort Diagram.
Figure 2. (A) Progression-free survival for all patients (n = 30) and (B) for LA (n = 22) and BR (n = 8) patients. (C) Overall survival curves of all patients (n = 30) and (D) LA (n = 22) and BR (n = 8) patients. (E) Overall survival for resected (n = 6) and non-resected (n = 24) patients.
Clinical studies of conventional CRT or GEM based chemotherapy for LAPC. Abbreviations: P II, phase II study; P III, phase III study; GEM, gemcitabine; CRT, chemoradiotherapy; RT, radiation therapy; PFS, progression-free survival; TTP, time to progression; MST, median survival time; NA, not available. a One-year progression-free survival.
Gemcitabine plus nab-paclitaxel for locally advanced or borderline resectable pancreatic cancer

November 2019

·

110 Reads

·

21 Citations

Overall survival in a phase III study for metastatic pancreatic cancer has significantly improved with gemcitabine (GEM) plus nab-paclitaxel. However, to date, there is limited data on the efficacy and safety of its use for patients with locally advanced (LA) or borderline resectable pancreatic cancer (BRPC). Here, we investigated the efficacy and safety of first-line GEM plus nab-paclitaxel for LA or BRPC. We retrospectively analysed consecutive patients with pathologically confirmed, untreated LA or BRPC who started receiving first-line GEM plus nab-paclitaxel. A total of 30 patients (LA, n = 22; BRPC, n = 8) were analysed. Twelve patients (40%) without distant metastasis received additional chemoradiotherapy using S-1. Laparotomy was performed on 8 patients and 6 (20%; LA, n = 3; BR, n = 3) achieved R0 resection. Objective response rate was 44.8%. For all patients, median progression-free survival and overall survival were 14.8 and 29.9 months, respectively. Median overall survival for LA was 24.1 months with a 2-year survival rate of 50.8%. The most frequently observed grade 3 or 4 toxicities were neutropenia (73%) and biliary infection (13%). First-line GEM plus nab-paclitaxel was well-tolerated and feasible with an encouraging survival for LA or BRPC.


Whole-exome sequencing in unresectable pancreatic cancer patients with long-term survival.

February 2019

·

41 Reads

Journal of Clinical Oncology

260 Background: Advances in technology of genomic sequencing have revealed the mutational landscape of pancreatic cancer. However, little is known with regard to molecular mechanisms underlying clinical diversity in “unresectable” advanced pancreatic cancer. Methods: We performed whole-exome sequencing using frozen cancer tissues prospectively obtained by EUS-FNA from primary tumors before chemotherapy in patients with metastatic or locally advanced pancreatic cancer (n = 35). All patients had pathological confirmation (adenocarcinoma, n = 34; carcinoma, n = 1). Somatic alterations in the cancer genome were compared with clinical outcomes. Results: Seventeen patients had metastatic disease and 18 had locally advanced disease. Thirty-four patients received chemotherapy with or without radiotherapy and one patient was treated with supportive care alone because of rapidly progressive disease. With a median follow-up time of 60.6 months (range 41.9–94.0) for censored cases, the median survival for all patients was 10.8 months. Nine patients survived more than 2 years ( > 6 years, n = 2; > 3 years, n = 3; > 2 years, n = 4), while 11 patients died within 4 months. We show mutational landscape of unresectable advanced pancreatic cancer and identified somatic mutations in known cancer related genes including KRAS (89%), TP53 (71%), SMAD4 (20%), CDKN2A (17%) and ARID1A (14%). We found that ARID1A mutation was mutually exclusive with TP53 mutation except for one tumor and had a significant correlation with survival outcomes. Among 9 patients who survived more than 2 years, 5 patients (56%) had ARID1A somatic mutation, whereas none (0%) had mutations in the remaining 26 patients who died within 2 years ( P = 0.0004). The median overall survival was 47.7 months for 5 patients with ARID1A-mutated tumors and 8.9 months for 30 patients with ARID1A wild-type tumors ( P = 0.0101). Conclusions: This is the first study to perform whole-exome sequencing in unresectable pancreatic cancer patients including very long-term survivors. We found that ARID1A mutations were associated with longer survival.


Mutational landscape of pancreatic cancer and genomic analysis using EUS-FNA samples

December 2018

·

10 Reads

·

1 Citation

Suizo

Advances in next-generation sequencing (NGS) technology have revealed the landscape of genomic alterations in patients with pancreatic cancer. NGS-based genomic profiling forms the basis of precision cancer medicine through the identification of potentially actionable mutations. However, few studies have addressed genomic analysis in patients with unresectable pancreatic cancer partly because of the difficulty in acquiring a biopsy sample suitable for genomic analysis due to abundant fibrosis in cancer tissue. In this article, we provide an overview of genomic alterations in patients with pancreatic cancer and discuss the current status and future issues regarding genomic analysis using EUS-FNA samples.



Study schematic
a Progression-free survival and b overall survival in the intention-to-treat population. c Overall survival of patients treated with CRT (n = 23) and those who did not receive planned CRT (n = 7)
Phase II study of induction gemcitabine and S-1 followed by chemoradiotherapy and systemic chemotherapy using S-1 for locally advanced pancreatic cancer

July 2017

·

27 Reads

·

13 Citations

Cancer Chemotherapy and Pharmacology

Purpose: S-1 has systemic activity for locally advanced pancreatic cancer (LAPC). Here, the efficacy and safety of induction gemcitabine (GEM) and S-1 (GS) followed by chemoradiotherapy (CRT) and systemic chemotherapy using S-1 for LAPC were assessed. Methods: The treatment consisted of four cycles of induction GS (S-1 60, 80, or 100 mg/day based on body surface area for 14 days every 3 weeks plus GEM 1000 mg/m(2) on days 8 and 15), followed by S-1 (80, 100, or 120 mg/day based on body surface area on days 1-14 and 22-35) and concurrent radiotherapy (50.4 Gy in 28 fractions). Maintenance chemotherapy with S-1 was started 1-4 weeks after CRT until disease progression or unacceptable toxicity was observed. The primary endpoint was 1-year survival. Results: A total of 30 patients with LAPC were enrolled. The median survival and progression-free survival were 21.3 and 12.7 months, respectively. Overall survival rates at 1, 2, 3, and 4 years were 73.3, 36.7, 23.3, and 16.7%, respectively. The median survival of 23 patients who received CRT was 22.9 months, with a 3-year survival rate of 30.4%. The two most common grade 3 or 4 adverse events during induction GS were neutropenia (63.3%) and biliary tract infection (20%). Toxicities during CRT or maintenance chemotherapy were generally mild. Conclusions: This regimen was feasible and highly active resulting in encouraging survival in patients with LAPC. Further investigations are warranted to elucidate the effectiveness of this treatment strategy in future studies. Clinical trials information: UMIN000006332.





Citations (23)


... Li and colleagues constructed a signetring cell organoid line from colon cancer, which recapitulated the histology of the original cancer tissue [25]. Hoshi and colleagues constructed an organoid line of acinar cell carcinoma from the pancreas, which could tolerate cryopreservation and recovery and showed functions similar to the original tissue [26]. However, there have been no reports on the construction of stable organoid lines from gastric cancer. ...

Reference:

Paired organoids from primary gastric cancer and lymphatic metastasis are useful for personalized medicine
Derivation of Pancreatic Acinar Cell Carcinoma Cell Line HS ‐1 as a Patient‐derived Tumor Organoid

... The effect of gemcitabine was also tested in two other pair-wise comparisons. A comparison between regimens that used single agent 5-FU [8,[13][14][15][16][17][18][19][20][21] to regimens that used [20,38,[67][68][69][70][71][72][73][74][75][76][77][78][79][80][81] showed a statistical trend toward improved OS (p = 0.017), and statistically significant improved PFS (p = 0.001) with the combination regimen. However, there was no significant difference in RR (p = 0.17). ...

A phase II trial of oral S-1 combined with gemcitabine in patients with unresectable biliary tract cancer
  • Citing Article
  • May 2009

Journal of Clinical Oncology

... Unfortunately, the response rate (RR) of gemcitabine monotherapy was found to be as low as 10%, meaning that only 1 out of every 10 patients would respond to treatment and see a shrinkage in tumor size [10]. When gemcitabine was paired with taxanes like nab-paclitaxel which work to arrest the cell cycle, its efficacy was drastically increased to 23% making it the new standard of care [11]. Although the combined therapy of gemcitabine and nab-paclitaxel was a major discovery in the fight against pancreatic cancer, better patient outcomes have been observed with other chemotherapy agents. ...

Gemcitabine plus nab-paclitaxel for locally advanced or borderline resectable pancreatic cancer

... Although the POLO study demonstrated that progression-free survival (PFS) was longer with olaparib maintenance than with placebo (7.4 vs. 3.8 months) among patients with metastatic pancreatic cancer and a germline BRCA mutation, PARP inhibitors were less commonly used than chemotherapy agents in the maintenance setting due to the low proportion of germline BRCA mutation (8). A phase II study showed that induction gemcitabine and S−1 (GS) followed by chemoradiotherapy and S−1 maintenance for locally-advanced disease was feasible, well-tolerated, and highly active, with median survival of 22.9 months and 3-year survival rate of 30.4% (9). Maintenance with capecitabine for metastatic disease treated with first-line FOLFIRINOX was also effective, with median OS (mOS) of 17 months and first PFS of 5 months (10). ...

Phase II study of induction gemcitabine and S-1 followed by chemoradiotherapy and systemic chemotherapy using S-1 for locally advanced pancreatic cancer

Cancer Chemotherapy and Pharmacology

... Changes in tumour blood perfusion, which can be detected as early as one or two weeks after the start of therapy, have been shown to differentiate between tumours that do or do not respond to chemoradiation [1], [2]. This is a much shorter time in comparison to the time it takes for reliable changes in tumour size-based biomarkers to be detected by conventional imaging methods, such as CT and MRI. ...

Changes in tumor vascularity depicted by contrast-enhanced ultrasonography as a predictor of chemotherapeutic effect in patients with unresectable pancreatic cancer
  • Citing Article
  • January 2009

Suizo

... Compared with the head of the pancreas, this risk is larger for tumors in the body and tail of the organ. [11][12][13] During the Whipple's treatment for lesions involving the head of the pancreas, the tract and the tumor were both removed, whereas the transgastric tract was not removed during distal pancreatectomy for tumors located in body and tail region. Two of our patients underwent EUS FNA and IHC of non-diagnostic lesions, which confirmed the diagnosis of SPN preoperatively. ...

A case of needle tract seeding after EUS-guided FNA in pancreatic cancer, detected by serial positron emission tomography/CT
  • Citing Article
  • February 2016

Gastrointestinal Endoscopy

... Generally, a few cases with signet ring cell carcinoma of eCCA were reported up to now (145). Previous studies also described two separable types containing intestinal type and pancreatobiliary type with CK7 negative plus CK20/MUC2 positive and CK7 positive plus CK20/MUC2 negative, respectively (146,147). That signet ring cell carcinoma of eCCA with distant lymph node metastasis has also been noticed recently (145). ...

Signet Ring Cell Carcinoma of the Extrahepatic Bile Duct Diagnosed by Preoperative Biopsy: A Case Report

Case Reports in Gastroenterology

... Gimeracil inhibits dihydropyrimidine dehydrogenase and prevents the breakdown of 5-FU, thereby prolonging its residence time in the body and enhancing the drug's efficacy. Oteracil reduces the activity of 5-FU in normal intestinal tissues by blocking its phosphorylation, thereby reducing the damage to normal tissues and minimizing the occurrence of adverse events (25)(26)(27). S-1 was associated with a lower rate of febrile neutropenia, toxicity-related deaths, grade 3-4 stomatitis, and mucositis than 5-fluorouracil (28). S-1, an oral medication, may help prevent AEs associated with intravenous injections. ...

S-1 in the treatment of pancreatic cancer

World Journal of Gastroenterology

... On the basis of a comparison with previous studies with GS or GnP 5,7,[10][11][12]28,29 , the OS rates of 17.9 months with GS and 13.3 months with GnP in the present study were among the best results for patients with advanced PDAC. The longer OS in both the GS and GnP groups might have been due to the inclusion of 28 (33.3%) ...

Randomized controlled study of gemcitabine plus S-1 combination chemotherapy versus gemcitabine for unresectable pancreatic cancer
  • Citing Article
  • December 2013

Cancer Chemotherapy and Pharmacology

... Ghrelin has been reported to decrease in the plasma of patients undergoing chemotherapy (Hiura et al., 2012a;Matsumura et al., 2013). A phase 2 clinical study examined the benefits of using ghrelin with ramosetron against acute emesis induced by cisplatin-based chemotherapy (Hiura et al., 2012b). ...

Changes in Plasma Ghrelin and Serum Leptin Levels after Cisplatin-Based Transcatheter Arterial Infusion Chemotherapy for Hepatocellular Carcinoma

ISRN Gastroenterology