Xanthine oxidoreductase and chemosensitivity in non-small cell lung cancer

2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 11077-11077
Author(s):  
R. Myint ◽  
M. Batus ◽  
P. Bonomi ◽  
P. Gattuso ◽  
W. H. Warren ◽  
...  

11077 Background: Xanthine oxidoreductase (XOR) is an enzyme involved in the degradation of purines into uric acid and reactive oxygen species and activation of the MAP kinase pathway involved in apoptosis. Decreased XOR expression was shown in recent studies to be associated with more aggressive disease in breast (Linder et al. Clin Cancer Res. 2005;11:4372–4381) and gastric cancers (Linder et al. J Clin Pathol. 2006;59:965–971). The goal of our study was to show that decreased XOR expression was associated with decreased survival in non-small cell lung cancer (NSCLC). Methods: Tissue specimens from 82 patients (pts) were stained using a XOR specific antibody (36 male and 46 female, age range from 40 to 92 years). These included 41 adenocarcinoma, 31 squamous cell, 8 poorly/moderately differentiated, and 2 bronchioloalveolar. XOR staining intensity was measured on a scale of 0 through 4 (0 being no staining). XOR intensity was correlated with clinical characteristics and outcomes using log rank and COX PH regression analysis. Results: Of the 82 pts, 34 received adjuvant chemo, and of these, 15 specimens had low XOR intensity (0–1). These 15 pts received adjuvant chemo and had a median survival of 543 days. In comparison, 19 of the 34 pts receiving adjuvant chemo had specimens with high XOR intensity (2–4). Their median survival was significantly longer at 2,023 days (p=0.007, hazard ratio=0.33). Conclusions: Although we had a small sample size, in our retrospective study, we found that pts who received adjuvant chemo had a longer survival if their tumors expressed high levels of XOR. XOR could be a potential predictor for responsiveness to adjuvant chemo in patients with NSCLC. Pts with decreased XOR may be less responsive to chemo and thus be able to avoid a toxic treatment if it is not significantly beneficial. [Table: see text] No significant financial relationships to disclose.

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 9548-9548
Author(s):  
James William Welsh ◽  
Dawei Chen ◽  
Paul Baas ◽  
Joe Y. Chang ◽  
Vivek Verma ◽  
...  

9548 Background: In metastatic non-small cell lung cancer (mNSCLC), the clinical trials NCT02492568 and NCT02444741 are the only known randomized comparisons of pembrolizumab alone versus pembrolizumab combined with radiation therapy (RT). When the trials were analyzed individually, some potential benefit was observed in the combination therapy group, but the relatively small sample size of each trial limited the detection of potential differences in response rates and outcomes. Hence, we perform a pooled analysis of these two randomized trials to validate and explore whether RT improves mNSCLC patient responses to immunotherapy. Methods: This was a pooled analysis of two randomized trials (NCT02492568 and NCT02444741) of pembrolizumab with or without RT for mNSCLC. Endpoints included the out-of-field overall response rate (ORR) and disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and subgroup analysis of the different RT schemes. Results: In all, 131 patients were analyzed (n = 66 pembrolizumab; n = 65 pembrolizumab/RT (iRT)). ORR was 21% in the pembrolizumab arm vs. 38% in the iRT arm (p = 0.01); DCR was 53% in the pembrolizumab arm vs. 67% in the iRT arm (p = 0.0009); PFS was 4.4 m vs 8.3 m (p = 0.046); and OS was 9.2 m vs 19.2 m (HR 0.66; p = 0.040). Ablative RT (24Gy/3 fractions and 50Gy/4 fractions) had better ORRs of 48% and 54%, respectively, compared to 18% for non-ablative RT (45Gy/15 fractions) and 20% for pembrolizumab alone (p < 0.05, respectively). Conclusions: The addition of RT to immunotherapy significantly increased the ORR of unirradiated lesions and was additionally associated with significant improvements in PFS and OS. Ablative RT was associated with response rates significantly higher than those of non-ablative RT, possibly due to a detrimental effect of non-ablative RT on ALC. These hypothesis-generating findings require dedicated, large-volume, and randomized studies for corroboration. Clinical trial information: NCT02492568 and NCT02444741 .


2022 ◽  
Vol 29 (1) ◽  
pp. 221-230
Author(s):  
Michael C. Tjong ◽  
Malavan Ragulojan ◽  
Ian Poon ◽  
Alexander V. Louie ◽  
Susanna Y. Cheng ◽  
...  

Background: The safety impact of radiotherapy (RT) timing relative to immune checkpoint inhibitors (ICIs) for advanced non-small-cell lung cancer (NSCLC) is unclear. We investigated if RT within 14 days (Interval 1) and 90 days (Interval 2) of ICI use is associated with toxicities compared to RT outside these intervals. Methods: Advanced NSCLC patients treated with both RT and ICIs were reviewed. Toxicities were graded as per CTCAE v4.0 and attributed to either ICIs or RT by clinicians. Associations between RT timing and Grade ≥2 toxicities were analyzed using logistic regression models adjusted for patient, disease, and treatment factors (α = 0.05). Results: Sixty-four patients were identified. Twenty received RT within Interval 1 and 40 within Interval 2. There were 20 Grade ≥2 toxicities in 18 (28%) patients; pneumonitis (6) and nausea (2) were most prevalent. One treatment-related death (immune encephalitis) was observed. Rates of patients with Grade ≥2 toxicities were 35%/25% in the group with/without RT within Interval 1 and 30%/25% in the group with/without RT within Interval 2. No significant association between RT timing relative to ICI use period and Grade ≥2 toxicities was observed. Conclusion: Albeit limited by the small sample size, the result suggested that pausing ICIs around RT use may not be necessary.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 7149-7149 ◽  
Author(s):  
D. Wang ◽  
A. Hanbali ◽  
M. Jankowski ◽  
E. U. Duran ◽  
A. Syed ◽  
...  

7149 Background: Hypercholesterolemia is one of the most common morbidities in United States. Statins have been proven as effective cholesterol-lowering agents and have been widely used in this country. They have been reported reducing the incidence and risk of various cancer types. However, the impact of using statins on the survival of patients with diagnosed non-small cell lung cancer is unknown. Methods: Ongoing retrospective study involves a cohort of 1,233 patients with diagnosed non-small cell lung cancer. Patients were identified from the Tumor Registry at Henry Ford Health System between January 1999 and December 2004. To date, the electronic medical records from 407 of the 1,233 cases were reviewed. Statistical analyses were performed and stratified for statin users versus non statin users. Results: Data from 407 patients were included in this analysis. There were 109 patients with hypercholesterolemia, 54 of them were statin users at the time of their non-small cell lung cancer diagnosis. Average age was 70 (range 46–85) years for statin users and 65 (range 29–93) years for non statin users. Median survival of 407 subjects was 9.82 months. After stratifying for statin use, the median survival of statin users was 12.38 months while the median survival of non statin users was 9.75 months, with a p value of 0.012. Conclusion: Though small sample size, this study is the first time ever suggesting a possible survival benefit of statin use in patients with non-small cell lung cancer who have received their standard anti-tumor regimens. Other variables that may contribute to this result will be further clarified and discussed after completing the data collection and analysis of this entire cohort. Based on our preliminary result, a prospective study of co-morbidity management with statins in patients with non-small cell lung cancer is warranted. No significant financial relationships to disclose.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e15055-e15055
Author(s):  
Richie Uba ◽  
Luis E. Raez ◽  
Katerine Dumais ◽  
Frank Gentile ◽  
Herman W. Powery ◽  
...  

e15055 Background: There is immune evasion and resistance to checkpoint inhibitors (CPI). Programmed death-ligand 1 (PD-L1) and Tumor mutational burden (TMB) might help us predict response, but we have not yet validated biomarkers that can predict resistance. Mutations (mut) in STK11 can induce epigenetic changes that confer proliferative advantages to cancer cells and preliminary reports have suggested that they can confer resistance to CPI. We investigated the role of STK11 and KRAS mut as markers of poor response to CPI in patients (pts) with non-small cell lung cancer (NSCLC). Methods: Clinical outcomes of 127 pts with stage IIIB-IV NSCLC who were tested for KRAS and STK11 mut and received CPI were evaluated for progression free survival (PFS) and overall survival (OS). For statistical analysis log-rank tests were used to compare OS and PFS, chi-squared tests were used to compare 1-year survivals and proportions among different variables, and Kaplan-Meier survival curves were used to report OS and PFS. Results: Of the 127 pts: 31 had STK11 mut, 14 had STK11+KRAS mut (SKM group), and 10 pts were in the SKMP group (STK11+KRAS mut+PD-L1 (-)). Median age was 65y (27-88y). Males were 54% of the total and 30 pts (24%) were Hispanic (H). STK11 mut patients had an inferior PFS and OS as shown in table. Pts in SKM and SKMP groups had worse outcomes; however, not all the P values were significant. The difference in OS and 1-year OS were very impressive and significant when compared between the SKMP group (4m and 30%) and the wild type group (15m and 73%). There were no significant differences in clinical outcomes for H vs. non-H White pts. Conclusions: The presence of STK11 mut were associated with shorter OS/PFS in NSCLC pts treated with CPIs, proving its utility as a negative predictive marker. This can be enhanced combining STK11 and KRAS mut and possibly adding PD-L1 (-). These findings are consistent with recent studies that have reported STK11 mut as a genomic driver of primary resistance. Due to the small sample size further studies will be needed to validate these findings. [Table: see text]


1987 ◽  
Vol 5 (5) ◽  
pp. 703-709 ◽  
Author(s):  
D H Johnson ◽  
M J DeLeo ◽  
K R Hande ◽  
S N Wolff ◽  
J D Hainsworth ◽  
...  

To exploit possible dose-response and combination drug synergism, 20 previously untreated patients with extensive-stage small-cell lung cancer (SCLC) received one or two courses of high-dose induction chemotherapy consisting of cyclophosphamide (100 mg/kg), etoposide (1,200 mg/m2), and cisplatin (120 mg/m2) (HDCEP). HDCEP was followed by four cycles of standard-dose cyclophosphamide (1,000 mg/m2), doxorubicin (40 mg/m2), and vincristine (1.4 mg/m2) (CAV). Response was determined after HDCEP and following CAV. Reevaluation included repeat bronchoscopy and chest computerized tomography (CT), as well as repetition of all initially abnormal studies. All patients were evaluable for response and toxicity. Overall response to HDCEP was 90%, with a complete response (CR) rate of 65% (95% confidence limits, 44% to 86%) and a partial response (PR) rate of 25% (95% confidence limits, 6% to 44%). All patients either maintained or improved their initial response while receiving CAV. Median duration of response was 6 months (range, 2 to 12 months) and median survival was 9.5 + months (range, 2 to 21 + months). All 37 courses of HDCEP were associated with leukopenia (less than 1,000/microL), 92% with thrombocytopenia (less than 20,000/microL), and 84% with fever of greater than 38.5 degrees C. Additional toxicities included bacteremia (24%), nausea and emesis (59%), mucositis (57%), diarrhea (38%), and hemorrhagic cystitis (5%). There were two treatment-related deaths due to infection. A third patient died 4 months after completing HDCEP with pulmonary fibrosis. Although response duration and median survival were not improved, HDCEP produced a high CR rate in ambulatory patients with extensive-stage SCLC.


Medicina ◽  
2009 ◽  
Vol 45 (6) ◽  
pp. 452 ◽  
Author(s):  
Saulius Cicėnas ◽  
Aurelija Žalienė ◽  
Vydmantas Atkočius

Objective. To determine survival of patients with stage IIIA/B non–small cell lung cancer considering disease stage and treatment methods. Material and methods. A total of 304 patients with non–small cell lung cancer were treated at the Department of Thoracic Surgery and Oncology, Institute of Oncology, Vilnius University, in 2000–2004. Stage IIIA (T3N1-2M0) cancer was diagnosed for 193 (63.5%) patients and stage IIIB (T4N0-1M0) cancer was diagnosed for 111 (36.5%) patients. There were 277 (91.1%) males and 27 (8.9%) females. According to morphology, there were 219 (72%) patients with squamous cell lung cancer, 80 (26.3%) with adenocarcinoma, and 5 (1.7%) patients with large cell carcinoma. Surgery was performed in 145 patients: 84 (57.9%) patients underwent lung resection (T3-4N0-1M0), 51 (35.2%) patients – thoracotomy, and 10 (6.7%) patients – other palliative thoracic procedures (mediastinotomy, pleurectomy, mediastinoscopy). Forty-eight (30.2%) patients were treated with radiation therapy with total doses of >40 Gy and 58 (36.5%) patients were treated with radiation therapy with total doses of <40 Gy. Fifty-four (33.9%) patients were treated with Gemzar and cisplatin and 19 (11.9%) patients were treated with etoposide and cisplatin.Results. Overall median and mean survival was 7.8 months (95% CI, 6.8 to 8.8) and 9.9 months (95% CI, 9.0 to 10.9), respectively. The median and mean survival of patients with stage IIIA cancer was 8.3 months and 10.4 months, respectively, and that of patients with stage IIIB cancer – 6.4 months and 9.0 months, respectively (P≤0.05). The median survival of the patients with stage IIIA cancer who received a combination of operation, chemotherapy, and radiation therapy with a total dose of >40 Gy was 14.4 months (mean, 14.7 months), and the median survival of those who received operation, chemotherapy, and radiation therapy with a total dose of ≤40 Gy was 9.7 months (mean, 14.1 months); the median survival of the patients who underwent surgery alone was 4.9 months (mean, 6.7 months) (P=0.004 and P=0.007), respectively. There was a significant difference in the median survival comparing the patients with stage IIIB cancer who underwent surgery alone and those who received a combination of radiation therapy and chemotherapy (median survival of 5.0 months [mean, 8.1 months] versus 16.8 months [mean, 17.6 months], respectively; P≤0.05). Conclusions. Disease stage had an influence on the survival of patients with non–small cell lung cancer: patients with stage IIIA (T3N0-1M0) cancer without metastases to mediastinal lymph nodes (N factor) survived longer than patients with stage IIIB (T4N1-2M0) cancer, where not only N factor had an impact but T factor as well. Better treatment outcomes, i.e. longer survival, can be achieved when a combination of three treatment types – surgery, chemotherapy, and radiation therapy – is applied to patients with stage IIIA or IIIB non–small cell lung cancer. The patients with stage IIIA disease who received surgery and radiation therapy (total dose, >40 Gy), and combinations of surgery, chemotherapy, and radiation therapy and second-line chemotherapy showed a significantly longer survival than those who received surgery alone.


1985 ◽  
Vol 3 (2) ◽  
pp. 176-183 ◽  
Author(s):  
H M Dhingra ◽  
M Valdivieso ◽  
D T Carr ◽  
D F Chiuten ◽  
P Farha ◽  
...  

One hundred sixty-seven evaluable patients with non-small-cell lung cancer were randomized to receive high-dose cisplatin and vindesine (PVD), or cisplatin and VP-16-213 (etoposide epipodophyllotoxin) (PVP), or cisplatin with VP-16-213 and vindesine (PVPVD). The patient distribution and characteristics were similar in all the treatment arms. The response rate differences (35% in PVD arm, 30% in PVP arm, and 22% in PVPVD arm) were not statistically significant (P = .33). Response durations were 43 weeks in the PVD arm, 20 weeks in the PVP arm, and 27 weeks in the PVPVD arm. Median survival was 29 weeks in the PVD and PVP arms and 28 weeks in the PVPVD arm. Median survival time of responding patients was 76 weeks in the PVD arm and 65 weeks in the PVP arm; 78% of patients were alive at 22+ to 87+ weeks follow-up in the PVPVD arm. Myelosuppression was similar in all three treatment arms. Significantly more azotemia occurred in the PVD arm than in the PVP and PVPVD arms (P = .002), and significantly more neuropathy in the PVD and PVPVD arms than in the PVP arm (P = .003 and .005). All the treatment arms have similar antitumor activity in non-small-cell lung cancer, but the PVP combination is slightly less toxic than the PVD and PVPVD treatment arms.


1988 ◽  
Vol 74 (2) ◽  
pp. 207-211
Author(s):  
Diego Tummarello ◽  
Francesco Guidi ◽  
Emilio Porfiri ◽  
Pierpaolo Isidor ◽  
Marzio Raspugli ◽  
...  

Sixty-one patients affected by small cell lung cancer (SCLC) entered in the study. Eighteen had limited disease and 43 extensive disease. Treatment consisted of: induction chemotherapy with 3 courses of CAV (cyclophosphamide, adriamycin, vincristine) in limited disease patients or 2 courses of CAV plus 2 courses of DDP-VP16 (cisplatin, etoposide) in extensive disease patients, followed by chest radiotherapy and CNS prophylaxis in responsive patients. Subsequently, responders and stable patients received maintenance chemotherapy by the alternation of cycles of CAV, DDP-VP16 and C'MP (CCNU, methotrexate, procarbazine), which lasted 1 year or until relapse. Four of 17 limited disease patients (23%) obtained a CR and 11 (65%) a PR; their median survival was 11 months (range, 2 + - 36 +). One of the 7 extensive disease patients (3%) achieved a CR and 19 (51%) a PR; their median survival was 6 months (range, 1-22). Median duration of response was 12 months for CR and 5 months for PR. Responders (CR and PR) survived 11.5 months versus 3.5 months for failures (P < 0.05); 3/61 (5%) showed long-term survival, in the absence of disease. The overall median survival was 7 months (range, 1-36 +). The main toxic effects were myelosuppression and vomiting (WHO grade 3). From our results, this program does not offer further substantial gains in patients with SCLC.


1990 ◽  
Vol 8 (1) ◽  
pp. 9-15 ◽  
Author(s):  
R T Chlebowski ◽  
L Bulcavage ◽  
M Grosvenor ◽  
E Oktay ◽  
J B Block ◽  
...  

This randomized, prospective, placebo-controlled clinical trial compares the influence on nutritional status and survival of hydrazine sulfate with placebo addition to cisplatin-containing combination chemotherapy in patients with unresectable non-small-cell lung cancer (NSCLC). The trial consisted of 65 patients with advanced, unresectable NSCLC who had had no prior chemotherapy, were at least partially ambulatory (Eastern Cooperative Oncology Group [ECOG] performance status [PS] level 0-2), and who had adequate hematologic, renal, and hepatic function. All patients received the same defined combination chemotherapy (cisplatin, vinblastine, and bleomycin) and the same defined dietary counseling with the addition of either three times daily oral hydrazine sulfate (60 mg) or placebo capsules. Hydrazine sulfate compared with placebo addition to chemotherapy resulted in significantly greater caloric intake and albumin maintenance (P less than .05). Considering all patients, survival was greater for the hydrazine sulfate compared with placebo group (median survival, 292 v 187 days), but the difference did not achieve statistical significance. In favorable PS patients (PS 0-1), survival was significantly prolonged (median survival, 328 days v 209 days; P less than .05) for hydrazine sulfate compared with placebo addition. In a multifactor analysis, PS, weight loss, and liver involvement were the final variables. Objective response frequency and toxicity were comparable on both arms. Hydrazine sulfate may favorably influence nutritional status and clinical outcome of patients with NSCLC. Further definitive studies of hydrazine sulfate addition to therapeutic regimens in NSCLC are warranted.


2009 ◽  
Vol 27 (26) ◽  
pp. 4254-4259 ◽  
Author(s):  
Bente Holm ◽  
Anders Mellemgaard ◽  
Torsten Skov ◽  
Birgit Guldhammer Skov

Purpose The excision repair cross-complementation group 1 (ERCC1) status was assessed in patients receiving carboplatin and gemcitabine for inoperable non–small-cell lung cancer (NSCLC). We analyzed the association between the ERCC1 status and the overall survival after the chemotherapy. Patients and Methods We retrospectively identified 163 patients with inoperable NSCLC and sufficient tumor tissue for ERCC1 analysis, who had received carboplatin and gemcitabine as first-line treatment. Immunohistochemistry was used to assess the expression of ERCC1. Results One hundred sixty-three patients were included. Seventy (42%) were ERCC1 positive. Patients treated with carboplatin and gemcitabine and having ERCC1-negative tumors had a significantly increased survival when compared to patients with ERCC1-positive tumors (median survival, 12.0 months v 8.2 months; P = .02). This difference was mainly seen in men, where those with ERCC1-negative tumors had a significantly increased survival compared to men with ERCC1-positive tumors (median survival, 11.8 months v 7.9 months; P = .005). Conversely, women who were ERCC1 negative did not have a survival advantage over ERCC1-positive women. Conclusion We confirmed previous reports that ERCC1 expression is predictive for outcome in patients treated with carboplatin and gemcitabine. Patients with ERCC1-negative tumors had an increased survival compared to patients with ERCC1-positive tumors and this difference was mainly attributable to a survival difference among men.


Sign in / Sign up

Export Citation Format

Share Document