Abstract B24: De novo and acquired resistance to first-line standard therapy in colorectal cancer: from cell lines to metastatic tumors

2012 ◽  
Vol 18 (10 Supplement) ◽  
pp. B24-B24
Author(s):  
Raquel Aloyz ◽  
Caroline Rousseau ◽  
Zuanel Diaz ◽  
Adriana Aguilar-Mahecha ◽  
Mark Basik ◽  
...  
2021 ◽  
Vol 22 (6) ◽  
pp. 3160
Author(s):  
Michal Kopczynski ◽  
Malgorzata Statkiewicz ◽  
Magdalena Cybulska ◽  
Urszula Kuklinska ◽  
Katarzyna Unrug-Bielawska ◽  
...  

TNF-related apoptosis-inducing ligand (TRAIL) is a type II transmembrane protein capable of selectively inducing apoptosis in cancer cells by binding to its cognate receptors. Here, we examined the anticancer efficacy of a recently developed chimeric AD-O51.4 protein, a TRAIL fused to the VEGFA-originating peptide. We tested AD-O51.4 protein activity against human colorectal cancer (CRC) models and investigated the resistance mechanism in the non-responsive CRC models. The quantitative comparison of apoptotic activity between AD-O51.4 and the native TRAIL in nine human colorectal cancer cell lines revealed dose-dependent toxicity in seven of them; the immunofluorescence-captured receptor abundance correlated with the extent of apoptosis. AD-O51.4 reduced the growth of CRC patient-derived xenografts (PDXs) with good efficacy. Cell lines that acquired AD-O51.4 resistance showed a significant decrease in surface TRAIL receptor expression and apoptosis-related proteins, including Caspase-8, HSP60, and p53. These results demonstrate the effectiveness of AD-O51.4 protein in CRC preclinical models and identify the potential mechanism underlying acquired resistance. Progression of AD-O51.4 to clinical trials is expected.


Cells ◽  
2020 ◽  
Vol 9 (1) ◽  
pp. 219 ◽  
Author(s):  
Nuria Garcia-Carbonero ◽  
Javier Martinez-Useros ◽  
Weiyao Li ◽  
Alberto Orta ◽  
Nuria Perez ◽  
...  

KRAS mutation is a confirmed predictive biomarker for anti-EGFR monoclonal antibody therapy response for metastatic colorectal cancer. However, its prognosis impact and the predictive potential for first-line standard chemotherapy remains unclear. On the other hand, V600E mutation is the most frequent and studied mutation in the BRAF gene, and it has been associated with a poor outcome of patients and a low response to anti-EGFR treatment. Thus, the aim of this study is to evaluate the role of KRAS and BRAF mutations as prognosis factors and predictive biomarkers for 1st line standard chemotherapy in metastatic colorectal cancer. KRAS mutations and BRAF V600E mutations exhibited a poor outcome (p = 0.021 and p < 0.0001, respectively). Cox multivariate analysis showed that the presence of liver metastasis (HR = 1.595; 95% CI: 1.086–2.343; p = 0.017), KRAS mutation (HR = 1.643; 95% CI: 1.110–2.431; p = 0.013) and BRAF V600E mutation (HR = 5.861; 95% CI: 2.531–13.570; p < 0.0001) were statistically significant co-variables for progression-free survival. Interestingly, patients with KRAS mutations were associated with a poor response to first line standard chemotherapy (p = 0.008). In contrast, the BRAF V600E mutation did not have any impact on the first line standard chemotherapy response (p = 0.540). Therefore, in the present study, we provide new insight on the role of KRAS and BRAF, not only as prognosis biomarkers, but also as first line standard chemotherapy response biomarkers in metastatic colorectal cancer.


JAMA Oncology ◽  
2019 ◽  
Vol 5 (3) ◽  
pp. 343 ◽  
Author(s):  
Chiara Cremolini ◽  
Daniele Rossini ◽  
Emanuela Dell’Aquila ◽  
Sara Lonardi ◽  
Elena Conca ◽  
...  

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 797-797 ◽  
Author(s):  
Jason Stephansky ◽  
Katsuhiro Togami ◽  
Mahmoud Ghandi ◽  
Joan Montero ◽  
Nick vonEgypt ◽  
...  

Abstract SL-401 is a novel targeted therapy comprised of recombinant interleukin 3 (IL3) fused to a truncated diphtheria toxin (DT) payload. SL-401 delivers DT to cells expressing the IL3 receptor (CD123). After internalization, DT catalyzes ADP ribosylation of eukaryotic elongation factor 2 (eEF2), blocking protein synthesis and killing target cells. SL-401 is currently in clinical trials for CD123+ cancers, including acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm (BPDCN). Other than CD123 expression, the determinants of response are largely unknown. Our goal was to study mechanisms of de novo and acquired resistance to inform combination strategies and enhance the efficacy of SL-401. In 16 AML and BPDCN patients enrolled in a phase 1-2 trial, we did not see decreased expression of CD123 by flow cytometry during or after SL-401. To study alternative resistance mechanisms, we generated 3-6 independent SL-401 resistant clones from each of 4 CD123+ AML (THP1, NOMO1, EOL1) or BPDCN (CAL1) cell lines. We treated cells with the LD95 (lethal dose to 95%), retreating upon recovery. All lines developed &gt;2-3 log resistance to SL-401 within 28 days. All resistant clones maintained CD123 surface expression, consistent with what we observed in patients. Using confocal microscopy, we saw that a fluorescently tagged SL-401 was internalized equally in resistant and parental cells. SL-401 resistant cells were also resistant to full length DT, suggesting that the mechanism of resistance involved DT rather than IL3 binding/internalization. We performed whole transcriptome RNA-sequencing and whole exome sequencing (WES) on parental and SL-401 resistant cells. There were no recurrent acquired DNA mutations. However, in RNA-seq the most downregulated gene in 6 independent clones from 2 lines was DPH1 (FC -7.5, FDR&lt;0.0001). DPH1 is the first enzyme in a cascade that converts histidine 715 on eEF2 to diphthamide, the direct target for ADP ribosylation by DT. Decreased expression of DPH1 was confirmed in the 6 resistant clones by qRT-PCR, and in 3 clones from an additional line. Across 33 cell lines and subclones there was an inverse linear correlation between DPH1 level and SL-401 IC50 (P=0.0005). To validate this finding in patients, we performed paired RNA-seq on CD45+CD123+ sorted blasts from 2 AMLs pre & post 2 cycles of SL-401. Both patients' AMLs had reduced DPH1 after exposure to SL-401 (mean -2.1 fold). To determine if loss of DPH1 was sufficient to confer de novo resistance to SL-401, we generated THP1, NOMO1, and CAL1 cells stably expressing the Cas9 nuclease and transduced them with 1 of 4 DPH1-targeting or 2 non-targeting CRISPR sgRNAs co-expressing GFP. Four days after infection at titers that achieve ~20% GFP+ cells, we treated with the LD95 of SL-401. We observed a survival advantage for DPH1 sgRNA-transduced cells (2.9 fold increase in GFP+ cells, P&lt;0.0001) and decreased apoptosis measured by AnnexinV positivity, compared to GFP- cells in the same cultures. By contrast, there was no survival advantage for cells transduced with control sgRNAs. These data suggest that DPH1 loss is sufficient to mediate SL-401 resistance in AML and BPDCN. Pseudomonas toxin (PT) also targets eEF2 on diphthamide, and PT resistance in a B-ALL cell line has been associated with DPH1 loss via reversible promoter CpG DNA methylation (Hu, Leuk Res 2013). Therefore, we tested the DNA methyltransferase inhibitor azacitidine in combination with SL-401 and observed synergistic cytotoxicity, in naïve (combination index (CI) = 0.45; &lt;1 indicates synergy) and SL-401 resistant (CI = 0.55) cells. Most strikingly, 4-week pulsatile treatment with non-lethal "epigenetic" doses of azacitidine (300 nM 2d on/2d off) fully reversed SL-401 resistance in 6 CAL1 and THP1 clones that were insensitive at baseline (Figure). Controls grown in vehicle or with weekly SL-401 challenge showed no reversion, suggesting that azacitidine had a specific sensitizing effect. Restoration of SL-401 sensitivity was accompanied by an increase in DPH1 expression compared to resistant clones. In summary, we found that DPH1 is a biomarker of SL-401 activity and acquired resistance, and resistance is reversible by azacitidine. Based on these data, we have initiated a multicenter phase 1 trial of the combination of SL-401 and azacitidine in patients with AML or MDS (NCT03113643), with correlative laboratory studies designed to explore these hypotheses. Disclosures vonEgypt: Stemline Therapeutics: Employment. Lindsay: Stemline Therapeutics: Employment. Brooks: Stemline Therapeutics: Employment, Equity Ownership, Patents & Royalties. Lane: Stemline Therapeutics: Research Funding; N-of-one: Consultancy.


2018 ◽  
Vol 36 (4_suppl) ◽  
pp. TPS886-TPS886
Author(s):  
Hiromichi Maeda ◽  
Naoki Nagata ◽  
Takeshi Nagasaka ◽  
Koji Oba ◽  
Hideyuki Mishima ◽  
...  

TPS886 Background: The mechanisms underlying the acquired resistance of metastatic colorectal cancer (mCRC) against panitumumab treatment is not fully understood. The efficacy and safety of FOLFIRI with panitumumab as the second-line chemotherapy after failure of FOLFOX with panitumumab treatment has yet to be determined. To address these two points, a multicenter single-arm Phase II clinical trial is being conducted with evaluation of circulating tumor DNA (ctDNA). Methods: The major inclusion criterion is that the patient has refractory measurable tumor that has progressed after the first-line therapy with FOLFOX plus panitumumab. After registration, treatment with the FOLFIRI and panitumumab will be continued in 2-week cycles until disease progression, unacceptable toxicity and/or patients’ refusal. The primary endpoint for this study is six-month progression-free survival (PFS) rate, a simple surrogate endpoint of PFS. According to a clinical trial revealing the median PFS of 4.6 months for FOLFIRI alone and 6.4 months for panitumumab plus FOLFIRI treatment in RAS wild-type patients (Peeters et al. Clin Cancer Res. 2015; 21: 5469-79), we assume the threshold and expected 6-month PFS rate as 35% and 50%, respectively. Under the settings of one-sided alpha = 0.10 and power = 80%, the required sample size is 53 patients. The target number of cases in this study is 55 patients, considering a dropout rate of 5%. The secondary endpoint includes the tumor-related gene mutation status assessed by liquid biopsy. Primary tumor and/or metastatic site tissue samples will be collected by formalin-fixed paraffin-embedded specimens at the time of registration. Blood samples will be collected at 3 time-points: (1) before second-line treatment, (2) at 6 ± 2 weeks after initiation of the treatment protocol, and (3) after confirmation of acquired resistance to this second-line therapy. The multiple evaluation of ctDNA will provide the meaningful information concerning relationship between the tumor resistance against treatment and alterations in gene mutation status. Clinical trial information: UMIN000026817.


Membranes ◽  
2020 ◽  
Vol 10 (5) ◽  
pp. 91
Author(s):  
Katia Cortese ◽  
Silvia Marconi ◽  
Cinzia Aiello ◽  
Maria Cristina Gagliani ◽  
Serena Pilato ◽  
...  

Colorectal cancer (CRC) is one of the main causes of cancer-related death in developed countries. Targeted therapies and conventional chemotherapeutics have been developed to help treat this type of aggressive cancer. Among these, the monoclonal antibodies cetuximab (Cxm) and panitumumab specifically target and inactivate the signaling of ERBB1 (EGF receptor), a key player in the development and progression of this cancer. Unfortunately, these antibodies are effective only on a small fraction of patients due to primary or secondary/acquired resistance. However, as ERBB1 cell surface expression is often maintained in resistant tumors, ERBB1 can be exploited as a target to deliver other drugs. Liposomes and immunoliposomes are under intensive investigation as pharmaceutical nanocarriers and can be functionalized with specific antibodies. In this study, we first investigated the anti-cancer activity of a cell permeable tripeptide, leucine-leucin-norleucinal (LLNle), an inhibitor of gamma-secretase and proteasome, in three different CRC cell lines that express ERBB1. We formulated LLNle-liposomes and Cxm-conjugated LLNle-loaded liposomes (LLNle-immunoliposomes) and evaluated their efficacy in inhibiting cell survival. Despite similar pro-apoptotic effects of free LLNle and LLNle-liposomes, immunoliposomes-LLNle were significantly less effective than their unconjugated counterparts. Indeed, immunoliposomes-LLNle were readily internalized and trafficked to lysosomes, where LLNle was likely trapped and/or inactivated. In conclusion, we demonstrated that LLNle was readily delivered to CRC cell lines by liposomes, but immunoliposomes-LLNle failed to show significant anti-cancer activity.


Sign in / Sign up

Export Citation Format

Share Document