Ramesh K. Ramanathan's research while affiliated with Mayo Clinic and other places

What is this page?


This page lists the scientific contributions of an author, who either does not have a ResearchGate profile, or has not yet added these contributions to their profile.

It was automatically created by ResearchGate to create a record of this author's body of work. We create such pages to advance our goal of creating and maintaining the most comprehensive scientific repository possible. In doing so, we process publicly available (personal) data relating to the author as a member of the scientific community.

If you're a ResearchGate member, you can follow this page to keep up with this author's work.

If you are this author, and you don't want us to display this page anymore, please let us know.

Publications (504)


DNA content flow sorting of FFPE EOCRC tissues. (A) Diploid tumor EOCRC11 with 2.0N (G0/G1) peak 2 (P2) and 4.0N (G2/M) peak 3 (P3). (B) Aneuploid tumor EOCRC12 with diploid (G0/G1) peak 2 (P2), 4.0N (G2/M) peak 5 (P5) and two distinct aneuploid peaks (P3 and P4). Each peak from individual samples was collected during sorting.
Mutational landscape of EOCRC. (A) Oncomap summary of somatic lesions in ASCP genomes. (B) Distribution of mutation frequency across EOCRC samples. Blue arrow EOCRC12 AN1, red arrow EOCRC12AN2. Red dashed horizontal line mean number of mutations. Black vertical line.
EGFR expression in EOCRC tumors. Immunohistochemistry (IHC) staining for EGFR in tumors with (A, B) high (3 +) expression (EOCRC19 and EOCRC12), and (C, D) negative (0) expression (EOCRC5, EOCRC20). Images at 40 × collected with Aperio ImageScope.
Single base substitution (SBS) mutation signatures of EOCRC. (A) SBS 1, 5, and 6 (red arrows) are present, SBS10 (blue arrow) is absent. (B) SBS 5 is present (red arrow) in high and (C) absent (blue arrow) in low tumor mutation burden (TMB) EOCRC samples.
CNV profiles of EOCRC. (A) Diploid EOCRC3 with focal 10q23.31 PTEN deletion (blue arrow). (B) Aneuploid (3.2N) population sorted from EOCRC12 with focal 10q23.31 PTEN deletion (blue arrow). (C) Aneuploid (3.5N) population sorted from EOCRC17 with focal 5q34 deletion (blue arrow) and focal 18q11 deletion (red arrow).
Genomic landscape of diploid and aneuploid microsatellite stable early onset colorectal cancer
  • Article
  • Full-text available

April 2024

·

11 Reads

Scientific Reports

Yumei Zhou

·

·

Jun Chen

·

[...]

·

Although colorectal cancer (CRC) remains the second leading cause of cancer-related death in the United States, the overall incidence and mortality from the disease have declined in recent decades. In contrast, there has been a steady increase in the incidence of CRC in individuals under 50 years of age. Hereditary syndromes contribute disproportionately to early onset CRC (EOCRC). These include microsatellite instability high (MSI+) tumors arising in patients with Lynch Syndrome. However, most EOCRCs are not associated with familial syndromes or MSI+ genotypes. Comprehensive genomic profiling has provided the basis of improved more personalized treatments for older CRC patients. However, less is known about the basis of sporadic EOCRC. To define the genomic landscape of EOCRC we used DNA content flow sorting to isolate diploid and aneuploid tumor fractions from 21 non-hereditary cases. We then generated whole exome mutational profiles for each case and whole genome copy number, telomere length, and EGFR immunohistochemistry (IHC) analyses on subsets of samples. These results discriminate the molecular features of diploid and aneuploid EOCRC and provide a basis for larger population-based studies and the development of effective strategies to monitor and treat this emerging disease.

Download
Share

A Randomized Controlled Trial of Fluorouracil Plus Leucovorin, Irinotecan, and Oxaliplatin Combinations in Patients With Previously Untreated Metastatic Colorectal Cancer

July 2023

·

27 Reads

·

24 Citations

Journal of Clinical Oncology

Purpose: Three agents with differing mechanisms of action are available for treatment of advanced colorectal cancer: fluorouracil, irinotecan, and oxaliplatin. In this study, we compared the activity and toxicity of three different two-drug combinations in patients with metastatic colorectal cancer who had not been treated previously for advanced disease. Patients and methods: Patients were concurrently randomly assigned to receive irinotecan and bolus fluorouracil plus leucovorin (IFL, control combination), oxaliplatin and infused fluorouracil plus leucovorin (FOLFOX), or irinotecan and oxaliplatin (IROX). The primary end point was time to progression, with secondary end points of response rate, survival time, and toxicity. Results: A total of 795 patients were randomly assigned between May 1999 and April 2001. A median time to progression of 8.7 months, response rate of 45%, and median survival time of 19.5 months were observed for FOLFOX. These results were significantly superior to those observed for IFL for all end points (6.9 months, 31%, and 15.0 months, respectively) or for IROX (6.5 months, 35%, and 17.4 months, respectively) for time to progression and response. The FOLFOX regimen had significantly lower rates of severe nausea, vomiting, diarrhea, febrile neutropenia, and dehydration. Sensory neuropathy and neutropenia were more common with the regimens containing oxaliplatin. Conclusion: The FOLFOX regimen of oxaliplatin and infused fluorouracil plus leucovorin was active and comparatively safe. It should be considered as a standard therapy for patients with advanced colorectal cancer.


A phase 1/2, first-in-human trial of ZB131, a novel antibody targeting cancer-specific plectin (CSP) in advanced solid tumors.

June 2023

·

13 Reads

·

1 Citation

Journal of Clinical Oncology

3083 Background: Cancer-Specific Plectin (CSP) is a novel pro-tumorigenic target that is selectively expressed on the surface of tumors, while absent from benign tissue. CSP is highly expressed in many solid cancers, particularly pancreatic (PC), cholangiocarcinoma (CCA), and ovarian (OC). ZB131 (Zielbio), is a humanized anti-CSP IgG1 monoclonal antibody that binds specifically to CSP on the surface of tumor cells, rapidly internalizes and modulates key proliferative and cytoskeletal remodeling signaling pathways to decrease cancer cell growth and increase the recruitment of anti-tumor immune infiltrates. Methods: The dose escalation (3+3 design) and expansion are enrolling adult patients (pts) with advanced treatment-refractory solid tumors. ZB131 was administered IV over 60 minutes (without premedication) weekly. The starting dose of ZB131 is 0.3 mg/kg followed by 1,3, 9, 15 and 30 mg/kg. Primary objectives are safety, tolerability, and maximum tolerated dose/determination of Phase 2 dose. Secondary objectives are preliminary efficacy (RECIST v1.1), Pharmacokinetic (PK) and pharmacodynamic parameters in blood and paired tumor biopsies. Results: As of 31 Jan 2023, 24 pts had enrolled (9 PC, 6 CCA, 4 OC, 5 other). The median age was 59.5 yr (range, 35 – 84) with a median of 4 prior lines of therapy. The median ZB131 treatment duration is 5.3 weeks (range, 1 – 33). No dose-limiting toxicities were observed up to the dose of 30 mg/kg which is currently enrolling. Treatment-related adverse events (TRAEs) were grade 1/ 2: nausea (n=6), fatigue (n=5), anemia (n=3), diarrhea (n=2), flulike symptoms (n=2), and vomiting (n=2). Mild flu-like symptoms and nausea/vomiting were observed during or following infusion at doses of 9 mg/kg and above. Except for one episode of Gr 3 neutropenia at the 30 mg/kg dose, no other drug-related grade 3/4 TRAE’s were reported. Five pts had stable disease range, 12 – 33 weeks (2 PC, 1 CCA, 2 OC), with a 43% decrease in CA-125 observed in one OC pt. The PKs are linear and dose-proportional for all dose levels evaluated (estimated T ½ of 6-9 days). No antidrug antibodies have been detected. Conclusions: Interim data from dose-escalation of this first-in-class, anti CSP antibody, demonstrates good tolerability with encouraging signs of activity and target engagement in heavily pretreated pts. Results support further clinical evaluation of ZB131 in dose-expansion cohorts including OC and including combination therapy with gemcitabine. Clinical trial information: NCT 05074472 .


Phase I Open-Label Study Evaluating the Safety, Pharmacokinetics, and Preliminary Efficacy of Dilpacimab in Patients with Advanced Solid Tumors

July 2021

·

47 Reads

·

7 Citations

Molecular Cancer Therapeutics

Dilpacimab (formerly ABT-165), a novel dual-variable domain immunoglobulin, targets both delta-like ligand 4 (DLL4) and VEGF pathways. Here, we present safety, pharmacokinetic (PK), pharmacodynamic (PD), and preliminary efficacy data from a phase 1 study (trial registration ID: NCT01946074) of dilpacimab in patients with advanced solid tumors. Eligible patients ({greater than or equal to}18 years) received dilpacimab intravenously on days 1 and 15 in 28-day cycles at escalating dose levels (range, 1.25-7.5 mg/kg) until progressive disease or unacceptable toxicity. As of August 2018, 55 patients with solid tumors were enrolled in the dilpacimab monotherapy dose-escalation and dose-expansion cohorts. The most common treatment-related adverse events (TRAEs) included hypertension (60.0%), headache (30.9%), and fatigue (21.8%). A TRAE of special interest was gastrointestinal perforation, occurring in two patients (3.6%; one with ovarian and one with prostate cancer), and resulting in one death. The PK of dilpacimab showed a half-life ranging from 4.9-9.5 days, and biomarker analysis demonstrated that the drug bound to both VEGF and DLL4 targets. The recommended phase 2 dose for dilpacimab monotherapy was established as 3.75 mg/kg, primarily on the basis of tolerability through multiple cycles. A partial response was achieved in 10.9% of patients (including four of 16 patients with ovarian cancer). The remaining patients had either stable disease (52.7%), progressive disease (23.6%), or were deemed unevaluable (12.7%). These results demonstrate that dilpacimab monotherapy has an acceptable safety profile, with clinical activity observed in patients with advanced solid tumors.


Multi-Cohort Retrospective Validation of a Predictive Biomarker for Topoisomerase I Inhibitors

December 2020

·

24 Reads

·

2 Citations

Clinical Colorectal Cancer

Purpose Camptothecin (CPT) analogues topotecan and irinotecan specifically target topoisomerase I (topoI), and are used to treat colorectal, gastric and pancreatic cancer. Patient response rate for this class of drug varies from 10-30% and there is no predictive biomarker for patient stratification by response. Based on our understanding of CPT drug resistance mechanisms, we developed an IHC-based predictive test (P-topoI-Dx) to stratify the responder versus non-responder patient populations. Patients and mthods The retrospective validation studies included a training set of (n=79) and a validation cohort (n=27) of gastric cancer (GC) patients, and eight cohorts of colorectal cancer (CRC) patient tissue (n=176). Progression-free survival for 6 months was considered positive response to CPT-based therapy. FFPE slides were immunohistochemically stained with anti-phosphospecific topoI-Serine10 (topoI-pS10), quantitated and analyzed statistically. Results We determined a threshold of 35% percent positive staining as offering optimal test characteristics in GC. The GC (n=79) training set demonstrated 76.6% (64-86) sensitivity; 68.8% (41-88) specificity; positive predictive value (PPV) 92.5% (81-98); and negative predictive value (NPV) 42.3% (24-62). The GC validation set (n=27) demonstrated 82.4% (56-95) sensitivity and 70.0% (35-92) specificity. Estimated PPV and NPV were 82.4% (56-95) and 70.0% (35-92) respectively. In the CRC validation set (n=176), the 40% threshold demonstrated 87.5% (78-94) sensitivity; 70.0% (59-79) specificity; PPV 70.7% (61-79) and NPV 87.0 % (77-93). Conclusion The analysis of retrospective data from patients (n=275) provides clinical validity to our P-topoI-Dx IHC test to identify the individuals who are more likely to respond to topoI inhibitors.



FIGURE 1. Nomogram to predict the OS of chemotherapy-naive patients with MPC.
Univariable Analyses of Potential Prognostic Factors and Multivariable Cox Proportional Hazards Model to Predict Survival
Scoring System for the Primary Nomogram to Predict OS
Nomogram for Estimating Overall Survival in Patients With Metastatic Pancreatic Cancer

June 2020

·

54 Reads

·

10 Citations

Pancreas

Objectives: This analysis investigated nomogram use to evaluate metastatic pancreatic cancer prognosis. Methods: Thirty-four baseline factors were examined in the Metastatic Pancreatic Adenocarcinoma Clinical Trial (MPACT) (nab-paclitaxel plus gemcitabine vs gemcitabine) data set. Factors significantly (P < 0.1) associated with overall survival (OS) in a univariable model or with known clinical relevance were tested further. In a multivariable model, factors associated with OS (P < 0.1) were selected to generate the primary nomogram, which was internally validated using bootstrapping, a concordance index, and calibration plots. Results: Using data from 861 patients, 6 factors were retained (multivariable analysis): neutrophil-lymphocyte ratio, albumin level, Karnofsky performance status, sum of longest diameter of target lesions, presence of liver metastases, and previous Whipple procedure. The nomogram distinguished low-, medium-, and high-risk groups (concordance index, 0.67; 95% confidence interval, 0.65-0.69; median OS, 11.7, 8.0, and 3.3 months, respectively). Conclusions: This nomogram may guide estimates of the range of OS outcomes and contribute to patient stratification in future prospective metastatic pancreatic cancer trials; however, external validation is required to improve estimate reliability and applicability to a general patient population. Caution should be exercised in interpreting these results for treatment decisions: patient characteristics could differ from those included in the nomogram development.


Serious treatment-emergent adverse events of grade ≥ 3 con- sidered related to copanlisib or refametinib a
Phase Ib Trial of the PI3K Inhibitor Copanlisib Combined with the Allosteric MEK Inhibitor Refametinib in Patients with Advanced Cancer

April 2020

·

46 Reads

·

33 Citations

Targeted Oncology

Background Dual inhibition of PI3K and MAPK signaling is conceptually a promising anticancer therapy.Objective This phase 1b trial investigated the safety, maximum tolerated dose (MTD), recommended phase II dose, pharmacokinetics, tumor response, fluorodeoxyglucose positron emission tomography (FDG-PET) pharmacodynamics, and biomarker explorations for the combination of pan-PI3K inhibitor copanlisib and allosteric MEK inhibitor refametinib in patients with advanced solid tumors.Patients and methodsThis was an adaptive trial with eight dose cohorts combining dose escalation and varying schedules in repeated 28-day cycles. Patients received copanlisib (0.2–0.8 mg/kg intravenously) intermittently (days 1, 8, 15) or weekly (days 1, 8, 15, 22) each cycle, and refametinib (30–50 mg twice daily orally) continuously or 4 days on/3 days off. Patients with KRAS, NRAS, BRAF, or PI3KCA mutations were eligible for the expansion cohort.ResultsIn the dose-escalation (n = 49) and expansion (n = 15) cohorts, the most common treatment-emergent adverse events included diarrhea (59.4%), nausea, acneiform rash, and fatigue (51.6% each). Dose-limiting toxicities included oral mucositis (n = 4), increased alanine aminotransferase/aspartate aminotransferase (n = 3), rash acneiform, hypertension (n = 2 each), and diarrhea (n = 1). MTD was copanlisib 0.4 mg/kg weekly and refametinib 30 mg twice daily. No pharmacokinetic interactions were identified. Decreased tumor FDG uptake and MEK-ERK signaling inhibition were demonstrated during treatment. Best response was stable disease (n = 21); median treatment duration was 6 weeks.Conclusions Despite sound rationale and demonstrable pharmacodynamic tumor activity in relevant tumor populations, a dose and schedule could not be identified for this drug combination that was both tolerable and offered clear efficacy in the population assessed.Clinicaltrials.gov identifierNCT01392521.


Disposition of patients in both schedules and all cohorts of the phase 1 imalumab study. Enrolled patients were those who had signed an informed consent form. AE, adverse event; Q2W, every two weeks; QW, every week
Area under the curve and maximum plasma concentration of imalumab given every two weeks (schedule 1) and once weekly (schedule 2). Data plotted are individual patient pharmacokinetic parameter values (estimate) versus actual total body dose of imalumab. AUCINF_obs, area under the plasma concentration‐time curve from time zero (pre‐dose) extrapolated to infinity (mg*h/L); AUC(0‐t), area under the plasma concentration‐time curve from time zero (pre‐dose) to time ‘t’ (338 hours post‐dose for schedule 1 and 168 hours post‐dose for schedule 2) (mg*h/L); Cmax, maximum plasma concentration (mg/L); PK_DAY = 1, cycle 1 day 1); PK‐DAY = 15, cycle 2 day 15. Q2W, every two weeks; QW, every week. The legend shows the treatment schedule, treatment cohort, and imalumab dose level for each data point
Concentration‐time curves of imalumab in schedule 1 (doses every two weeks, A) and schedule 2 (doses every week, B). QW, weekly; Q2W, every two weeks. Error bars represent standard deviation
Tissue penetration (a) and target occupancy (B) showing imalumab binding to oxMIF in mCRC tumour tissue. (A) Example tissue penetration illustrated by stained regions: Consecutive slides were stained for imalumab and oxMIF. Target occupancy was calculated based on digital image analysis. (B) Target occupancy in a patient with mCRC: Median target occupancy in total tumour tissue (tumor + stroma) reached 102% after the first treatment cycle and increased to 119% after the second cycle. Patient was treated with 10 mg/kg imalumab, QW. C2D1, cycle 2, day 1; C2D28, cycle 2, day 28; mCRC, metastatic colorectal cancer; oxMIF, oxidized macrophage migration inhibitory factor; QW, weekly
Phase I study of imalumab (BAX69), a fully human recombinant antioxidized macrophage migration inhibitory factor antibody in advanced solid tumours

April 2020

·

190 Reads

·

41 Citations

British Journal of Clinical Pharmacology

British Journal of Clinical Pharmacology

Aim Preclinical evidence suggests that oxidized macrophage migration inhibitory factor (oxMIF) may be involved in carcinogenesis. This phase 1 study (NCT01765790) assessed the safety, tolerability, pharmacokinetics and antitumour activity of imalumab, an oxMIF inhibitor, in patients with advanced cancer using ‘3 + 3’ dose escalation. Methods In Schedule 1, patients with solid tumours received doses from 1 to 50 mg/kg IV every 2 weeks. In Schedule 2, patients with metastatic colorectal adenocarcinoma, non‐small‐cell lung, or ovarian cancer received weekly doses of 10 or 25 mg/kg IV (1 cycle = 28 days). Treatment continued until disease progression, unacceptable toxicity, dose‐limiting toxicity, or withdrawal of consent. Results Fifty of 68 enrolled patients received imalumab. The most common treatment‐related adverse events (TRAEs) included fatigue (10%) and vomiting (6%); four grade 3 serious TRAEs (two patients) occurred. The dose‐limiting toxicity was allergic alveolitis (one patient, 50 mg/kg every 2 weeks). The maximum tolerated and biologically active doses were 37.5 mg/kg every 2 weeks and 10 mg/kg weekly, respectively. Of 39 assessed patients, 13 had stable disease (≥4 months in 8 patients). Conclusions Imalumab had a maximum tolerated dose of 37.5 mg/kg every 2 weeks in patients with advanced solid tumours, with a biologically active dose of 10 mg/kg weekly. Further investigation will help define the role of oxMIF as a cancer treatment target.


Phase 1 Dose-Escalation and -Expansion Study of Telisotuzumab (ABT-700), an Anti-c-Met Antibody, in Patients With Advanced Solid Tumors

March 2020

·

41 Reads

·

17 Citations

Molecular Cancer Therapeutics

This first-in-human phase 1 study evaluated the pharmacokinetics, safety, and preliminary efficacy of telisotuzumab, formerly called ABT-700, an antagonistic antibody directed against c-Met. For dose-escalation (3+3 design), three to six patients with advanced solid tumors were enrolled into four dose cohorts (5-25 mg/kg). In the dose-expansion phase, a subset of patients were prospectively selected for MET amplification (FISH screening). Patients received telisotuzumab intravenously on day 1 every 21 days. For dose expansion, 15 mg/kg was chosen as the dose on the basis of safety, pharmacokinetics, and other data from the escalation cohorts. Forty-five patients were enrolled and received at least one dose of telisotuzumab (dose escalation, n=15; dose expansion, n=30). Telisotuzumab showed a linear pharmacokinetics profile; peak plasma concentration was proportional to dose level. There were no acute infusion reactions and no dose-limiting toxicities were observed. The most common treatment-related adverse events included hypoalbuminemia (n=9, 20.0%) and fatigue (n=5, 11.1%). By Response Evaluation Criteria In Solid Tumors (RECIST), four of 10 (40.0%) patients with MET-amplified tumors had confirmed partial response in target lesions (one ovarian, two gastric, and one esophageal), two (20.0%) had stable disease, three (30.0%) had progressive disease; one patient was unable to be evaluated. Among patients with nonamplified tumors (n=35), no objective responses were observed; however, 11 patients had stable disease per RECIST criteria. In conclusion, telisotuzumab has an acceptable safety profile with clinical activity observed in patients with MET-amplified advanced solid tumors.


Citations (72)


... USP3 forms a ceRNA network with miR-224, SMAD4 [67]. Oxaliplatin blocks DNA replication and transcription and is considered the standard drug for advanced CRC [68]. USP3 can also impact CRC sensitivity to oxaliplatin by mediating the USP3/AR/RASGRP3 axis. ...

Reference:

Ubiquitin specific peptidase 3: an emerging deubiquitinase that regulates physiology and diseases
A Randomized Controlled Trial of Fluorouracil Plus Leucovorin, Irinotecan, and Oxaliplatin Combinations in Patients With Previously Untreated Metastatic Colorectal Cancer
  • Citing Article
  • July 2023

Journal of Clinical Oncology

... It was developed by AbbVie and was used in a phase I dose-escalation study among patients with advanced solid tumors (NCT01946074). The preliminary efficacy data reported partial responses in 10.9% of the patients, with the remaining patients having either stable (52.7%) or progressive disease (12.7%) [92]. ...

Phase I Open-Label Study Evaluating the Safety, Pharmacokinetics, and Preliminary Efficacy of Dilpacimab in Patients with Advanced Solid Tumors

Molecular Cancer Therapeutics

... (4)(5)(6) For many years, iCCAs were joined together with other BTCs, and clinical trials have traditionally recruited all subgroups of BTC. In fact, current standard-of-care adjuvant (7,8) and palliative (9)(10)(11)(12) chemotherapy for BTC does not distinguish between BTC subtypes. In contrast, there is increasing evidence suggesting that iCCA, eCCA, GBC, and AMPs have different etiological, clinical, genomic, and molecular characteristics. ...

A phase II trial of gemcitabine (G), cisplatin (C), and nab-paclitaxel (N) in advanced biliary tract cancers (aBTCs).
  • Citing Article
  • May 2017

Journal of Clinical Oncology

... Emerging evidence supporting the importance of the stroma in PDAC led to the therapeutic approaches that deplete the stroma. Clinically (NCT01130142, NCT01959139), anti-CAF drugs reduced the stroma, but unexpectedly resulted in a more aggressive tumor phenotype, enhancement of tumor immune evasion, and development of chemoresistance [10,11]. These results emphasize the complex role of the stroma in balancing tumor-promoting and -restraining functions [12,13] and the necessity of non-cytotoxic, CAF-directed approaches. ...

A phase IB/II randomized study of mFOLFIRINOX (mFFOX) + pegylated recombinant human hyaluronidase (PEGPH20) versus mFFOX alone in patients with good performance status metastatic pancreatic adenocarcinoma (mPC): SWOG S1313 (NCT #01959139).
  • Citing Article
  • February 2018

Journal of Clinical Oncology

... P = 0.19; Fig. 2c). The median OS was 12.5 months for the Malignant history, n (%) Yes 34 (13) 19 (12) Malignant family history, n (%) Yes 75 (29) 48 (30) Previous tumor resection, n (%) Yes 44 (17) 20 (13) Previous biliary drainage, n (%) Yes 65 (26) 39 (25) Pancreatic tumor location, n (%) Head 123 (48) 75 (47) Body/tail 132 (52) 84 (53) Histology, n (%) Adenocarcinoma 212 (83) 138 (87) Others 10 (4) 3 (2) Unknown 33 (13) 18 (11) Site of metastatic disease, n (%) Liver 154 (60) 100 (63) Peritoneum 62 (24) 37 (23) Lung 39 (15) 22 (14) Number of metastatic sites, n (%) ≥ 2 97 (38) 59 (37) Ascites, n (%) Yes 56 (22) 29 (18) First-line chemotherapy, n (%) FFX 102 (40) In other regimens, the median OS for the CAR < 0.54 group was 5.2 months, whereas it was 2.9 months for the CAR ≥ 0.54 group. There was no significant difference between these two groups (HR 1.64; 95%CI 0.93-2.88; ...

Nomogram for Estimating Overall Survival in Patients With Metastatic Pancreatic Cancer

Pancreas

... Mek-PI3K signaling was inhibited, and tumor FDG absorption was decreased following this therapy for 6 weeks. This combination of MEK-PI3K inhibitors is being studied in phase Ib trials in patients with advanced cancer (Table 1, Figure 6) [52]. ...

Phase Ib Trial of the PI3K Inhibitor Copanlisib Combined with the Allosteric MEK Inhibitor Refametinib in Patients with Advanced Cancer

Targeted Oncology

... Imalumab increases apoptosis by suppressing MIF-induced phosphorylation of ERK1/2 and AKT. Imalumab also makes cancer cell lines more susceptible to cytotoxic medications [38]. Moreover, in clinical practice, serum MIF levels could be utilized to stratify patients based on the aggressiveness of their disease, potentially guiding treatment decisions. ...

Phase I study of imalumab (BAX69), a fully human recombinant antioxidized macrophage migration inhibitory factor antibody in advanced solid tumours
British Journal of Clinical Pharmacology

British Journal of Clinical Pharmacology

... Elevated MET activity has been observed across various malignancies, often leading to treatment resistance. While therapeutic strategies such as monoclonal antibodies (mAbs) have demonstrated the ability to stabilize the disease, they have not significantly extended patient survival in advanced cancer cases [37,38]. The future success of targeting the MET pathway hinges on innovative drug design strategies and the identification of biomarkers that can guide treatment decisions [36]. ...

Phase 1 Dose-Escalation and -Expansion Study of Telisotuzumab (ABT-700), an Anti-c-Met Antibody, in Patients With Advanced Solid Tumors
  • Citing Article
  • March 2020

Molecular Cancer Therapeutics

... A phase II study showed the safety of BMP31510/gemcitabine in patients with advanced PDAC. 73 GP-2250 is an oxathiazine derivative with the ability to inhibit GAPDH (glyceraldehyde-3-phosphate dehydrogenase), a rate-limiting enzyme in aerobic glycolysis. 74 It causes the production of reactive oxygen species and induces apoptosis preferentially in the cancer cells. ...

Phase II trial of BPM31510-IV plus gemcitabine in advanced pancreatic ductal adenocarcinomas (PDAC).
  • Citing Article
  • February 2020

Journal of Clinical Oncology

... 14 However, disappointingly, phase I and II studies of inhibitors targeting this signaling pathway, such as saridegib (IPI-926) or vismodegib (GDC-0449), failed to demonstrate any significant benefit for this strategy. 15,16 Similarly, degradation of hyaluronan, a major component of the pancreatic ductal adenocarcinoma extracellular matrix, was presented as a novel strategy to allow high concentrations of chemotherapy to reach the tumor, resulting in improved survival and revealing an underappreciated sensitivity of the disease to conventional cytotoxic agents in KPC murine models. 17 Encouraged by the preliminary data from phase I/II studies, 18,19 pegvorhyaluronidase alfa (PEGPH20), a PEGylated recombinant human hyaluronidase, was tested in larger randomized controlled trials, but demonstrated no efficacy in combination with nab-paclitaxel/gemcitabine. ...

Phase 2 study of vismodegib, a hedgehog inhibitor, combined with gemcitabine and nab-paclitaxel in patients with untreated metastatic pancreatic adenocarcinoma

British Journal of Cancer