Andrew Nixon
Overview:
Andrew Nixon, PhD, MBA (Professor of Medicine) is Director of the Phase I Biomarker Laboratory, which brings together clinical, translational and basic research to pursue the development of novel biomarkers defining mechanisms of sensitivity, resistance, and toxicity to given therapeutic drug classes, particularly anti-angiogenic agents. Additionally, the laboratory has been appointed as a Molecular Reference Laboratory for the Alliance oncology cooperative group, a national clinical trial research group sponsored by the National Cancer Institute. The laboratory has quality control procedures in place to address many of the issues involved in clinical trial research including determination of sample quantity, sample integrity, and sample heterogeneity. We have spent considerable time developing robust assays that utilize limited amounts of specimen while providing high quality data. Multiplex ELISA and gene expression arrays are used to analyze serially collected blood and paraffin samples archived from cancer patient clinical trials. This work has the potential to improve the efficacy and toxicity of current therapies and to guide the development of the next generation of anti-angiogenesis therapies for cancer and other diseases.
Positions:
Professor in Medicine
Medicine, Medical Oncology
School of Medicine
Member of the Duke Cancer Institute
Duke Cancer Institute
School of Medicine
Education:
Ph.D. 1997
Wake Forest University
M.B.A. 2007
Duke University
Grants:
Preclinical and Human Correlative Studies of a Novel Bruton Tyronsine Kinase Inhibitor in Pancreatic Cancer
Administered By
Medicine, Medical Oncology
Awarded By
Department of Defense
Role
Co Investigator
Start Date
End Date
Plasma Angiome and Serum Androgens as Predictors of Overall Survival in Metastatic Prostate Cancer
Administered By
Biostatistics & Bioinformatics
Awarded By
Department of Defense
Role
Partnering PI
Start Date
End Date
(PQA5) 'Dose and Mechanisms of Exercise in Breast Cancer Prevention'
Administered By
Radiation Oncology
Awarded By
National Institutes of Health
Role
Co Investigator
Start Date
End Date
Anti-VEGF in Tumors & Wounds: Efficacy vs Toxicity
Administered By
Medicine, Medical Oncology
Awarded By
National Institutes of Health
Role
Co Investigator
Start Date
End Date
Regulation Of Cyclic Gmp Phosphodiesterase By Gz
Administered By
Pharmacology & Cancer Biology
Awarded By
National Institutes of Health
Role
PI-Fellow
Start Date
End Date
Publications:
Supplementary Figure 4B from Angiokines Associated with Targeted Therapy Outcomes in Patients with Non–Clear Cell Renal Cell Carcinoma
<jats:p><p>Supplementary Figure 4B Kaplan-Meier OS curves by treatment and dichotomized angiokines groups for following angiokines: TGFb-R3, HGF, TSP-2, OPN, IL-6, SDF-1, Ang-2, ICAM-1, TGF-b2, TIMP-1, BMP-9, PDGF-BB, VCAM-1, PDGF-AA, VEGF-D, TGF-b1, VEGF-R1, HER-3, VEGF, PlGF, CD-73, VEGF-R3, and VEGF-R2</p></jats:p>
Authors
Armstrong, AJ; Nixon, AB; Carmack, A; Yang, Q; Eisen, T; Stadler, WM; Jones, RJ; Garcia, JA; Vaishampayan, UN; Picus, J; Hawkins, RE; Hainsworth, JD; Kollmannsberger, CK; Logan, TF; Puzanov, I; Pickering, LM; Ryan, CW; Protheroe, A; George, DJ; Halabi, S
MLA Citation
Armstrong, Andrew J., et al. Supplementary Figure 4B from Angiokines Associated with Targeted Therapy Outcomes in Patients with Non–Clear Cell Renal Cell Carcinoma. 31 Mar. 2023. Crossref, doi:10.1158/1078-0432.22478717.v1.
URI
https://scholars.duke.edu/individual/pub1572091
Source
crossref
Published Date
DOI
10.1158/1078-0432.22478717.v1
NIH SenNet Consortium to map senescent cells throughout the human lifespan to understand physiological health.
Cells respond to many stressors by senescing, acquiring stable growth arrest, morphologic and metabolic changes, and a proinflammatory senescence-associated secretory phenotype. The heterogeneity of senescent cells (SnCs) and senescence-associated secretory phenotype are vast, yet ill characterized. SnCs have diverse roles in health and disease and are therapeutically targetable, making characterization of SnCs and their detection a priority. The Cellular Senescence Network (SenNet), a National Institutes of Health Common Fund initiative, was established to address this need. The goal of SenNet is to map SnCs across the human lifespan to advance diagnostic and therapeutic approaches to improve human health. State-of-the-art methods will be applied to identify, define and map SnCs in 18 human tissues. A common coordinate framework will integrate data to create four-dimensional SnC atlases. Other key SenNet deliverables include innovative tools and technologies to detect SnCs, new SnC biomarkers and extensive public multi-omics datasets. This Perspective lays out the impetus, goals, approaches and products of SenNet.
Authors
SenNet Consortium,
MLA Citation
SenNet Consortium, Daxin. “NIH SenNet Consortium to map senescent cells throughout the human lifespan to understand physiological health.” Nat Aging, vol. 2, no. 12, Dec. 2022, pp. 1090–100. Pubmed, doi:10.1038/s43587-022-00326-5.
URI
https://scholars.duke.edu/individual/pub1561272
PMID
36936385
Source
pubmed
Published In
Nature Aging
Volume
2
Published Date
Start Page
1090
End Page
1100
DOI
10.1038/s43587-022-00326-5
Data from Predictive Biomarkers of Overall Survival in Patients with Metastatic Renal Cell Carcinoma Treated with IFNα ± Bevacizumab: Results from CALGB 90206 (Alliance)
<jats:p><div>AbstractPurpose:<p>CALGB 90206 was a phase III trial of 732 patients with metastatic renal cell carcinoma (mRCC) comparing bevacizumab plus IFNα (BEV + IFN) with IFNα alone (IFN). No difference in overall survival (OS) was observed. Baseline samples were analyzed to identify predictive biomarkers for survival benefit.</p>Patients and Methods:<p>A total of 32 biomarkers were assessed in 498 consenting patients randomly assigned into training (<i>n</i> = 279) and testing (<i>n</i> = 219) sets. The proportional hazards model was used to test for treatment arm and biomarker interactions of OS. The estimated coefficients from the training set were used to compute a risk score for each patient and to classify patients by risk in the testing set. The resulting model was assessed for predictive accuracy using the time-dependent area under the ROC curve (tAUROC).</p>Results:<p>A statistically significant three-way interaction between IL6, hepatocyte growth factor (HGF), and bevacizumab treatment was observed in the training set and confirmed in the testing set (<i>P</i> < 0.0001). The model based on IL6, HGF, and bevacizumab treatment was predictive of OS (<i>P</i> < 0.001), with the high- and low-risk groups having a median OS of 10.2 [95% confidence interval (CI), 8.0–13.8] and 34.3 (95% CI, 28.5–40.5) months, respectively. The average tAUROC for the final model of OS based on 100 randomly split testing sets was 0.78 (first, third quartiles = 0.77, 0.79).</p>Conclusions:<p>IL6 and HGF are potential predictive biomarkers of OS benefit from BEV + IFN in patients with mRCC. The model based on key biological and clinical factors demonstrated predictive efficacy for OS. These markers warrant further validation in future anti-VEGF and immunotherapy in mRCC trials.</p><p><i><a href="http://dx.doi.org/10.1158/1078-0432.CCR-22-0750" target="_blank">See related commentaries by Mishkin and Kohn, p. 2722</a> and <a href="http://dx.doi.org/10.1158/1078-0432.CCR-22-0027" target="_blank">George and Bertagnolli, p. 2725</a></i></p></div></jats:p>
Authors
Nixon, AB; Halabi, S; Liu, Y; Starr, MD; Brady, JC; Shterev, I; Luo, B; Hurwitz, HI; Febbo, PG; Rini, BI; Beltran, H; Small, EJ; Morris, MJ; George, DJ
MLA Citation
Nixon, Andrew B., et al. Data from Predictive Biomarkers of Overall Survival in Patients with Metastatic Renal Cell Carcinoma Treated with IFNα ± Bevacizumab: Results from CALGB 90206 (Alliance). 31 Mar. 2023. Crossref, doi:10.1158/1078-0432.c.6531354.v1.
URI
https://scholars.duke.edu/individual/pub1571456
Source
crossref
Published Date
DOI
10.1158/1078-0432.c.6531354.v1
Data from Combination of PARP Inhibitor Olaparib, and PD-L1 Inhibitor Durvalumab, in Recurrent Ovarian Cancer: a Proof-of-Concept Phase II Study
<jats:p><div>AbstractPurpose:<p>Preclinical studies suggest PARP inhibition (PARPi) induces immunostimulatory micromilieu in ovarian cancer thus complementing activity of immune checkpoint blockade. We conducted a phase II trial of PARPi olaparib and anti–PD-L1 durvalumab and collected paired fresh core biopsies and blood samples to test this hypothesis.</p>Patients and Methods:<p>In a single-center, proof-of-concept phase II study, we enrolled women aged ≥18 with recurrent ovarian cancer. All patients were immune checkpoint inhibitor–naïve and had measurable disease per RECISTv1.1, ECOG performance status 0–2, and adequate organ and marrow function. Patients received olaparib 300 mg twice daily and durvalumab 1,500 mg intravenously every 4 weeks until disease progression, unacceptable toxicity, or withdrawal of consent. Primary endpoint was overall response rate (ORR). Secondary objectives were safety and progression-free survival (PFS). Translational objectives included biomarker evaluation for relationships with clinical response and immunomodulatory effects by treatment.</p>Results:<p>Thirty-five patients with ovarian cancer [median, four prior therapies (IQR, 2–5.5), predominantly platinum-resistant (86%), <i>BRCA</i> wild-type (77%)] received at least one full cycle of treatment. ORR was 14% [5/35; 95% confidence interval (CI), 4.8%–30.3%]. Disease control rate (PR+SD) was 71% (25/35; 95% CI, 53.7%–85.4%). Treatment enhanced <i>IFNγ</i> and <i>CXCL9/CXCL10</i> expression, systemic IFNγ/TNFα production, and tumor-infiltrating lymphocytes, indicating an immunostimulatory environment. Increased IFNγ production was associated with improved PFS [HR, 0.37 (95% CI, 0.16–0.87), <i>P</i> = 0.023], while elevated VEGFR3 levels were associated with worse PFS (HR, 3.22 (95% CI, 1.23–8.40), <i>P</i> = 0.017].</p>Conclusions:<p>The PARPi and anti–PD-L1 combination showed modest clinical activity in recurrent ovarian cancer. Our correlative study results suggest immunomodulatory effects by olaparib/durvalumab in patients and indicate that VEGF/VEGFR pathway blockade would be necessary for improved efficacy of the combination.</p></div></jats:p>
Authors
MLA Citation
Lampert, Erika J., et al. Data from Combination of PARP Inhibitor Olaparib, and PD-L1 Inhibitor Durvalumab, in Recurrent Ovarian Cancer: a Proof-of-Concept Phase II Study. 31 Mar. 2023. Crossref, doi:10.1158/1078-0432.c.6530135.
URI
https://scholars.duke.edu/individual/pub1572092
Source
crossref
Published Date
DOI
10.1158/1078-0432.c.6530135
Data from Predictive Biomarkers of Overall Survival in Patients with Metastatic Renal Cell Carcinoma Treated with IFNα ± Bevacizumab: Results from CALGB 90206 (Alliance)
<jats:p><div>AbstractPurpose:<p>CALGB 90206 was a phase III trial of 732 patients with metastatic renal cell carcinoma (mRCC) comparing bevacizumab plus IFNα (BEV + IFN) with IFNα alone (IFN). No difference in overall survival (OS) was observed. Baseline samples were analyzed to identify predictive biomarkers for survival benefit.</p>Patients and Methods:<p>A total of 32 biomarkers were assessed in 498 consenting patients randomly assigned into training (<i>n</i> = 279) and testing (<i>n</i> = 219) sets. The proportional hazards model was used to test for treatment arm and biomarker interactions of OS. The estimated coefficients from the training set were used to compute a risk score for each patient and to classify patients by risk in the testing set. The resulting model was assessed for predictive accuracy using the time-dependent area under the ROC curve (tAUROC).</p>Results:<p>A statistically significant three-way interaction between IL6, hepatocyte growth factor (HGF), and bevacizumab treatment was observed in the training set and confirmed in the testing set (<i>P</i> < 0.0001). The model based on IL6, HGF, and bevacizumab treatment was predictive of OS (<i>P</i> < 0.001), with the high- and low-risk groups having a median OS of 10.2 [95% confidence interval (CI), 8.0–13.8] and 34.3 (95% CI, 28.5–40.5) months, respectively. The average tAUROC for the final model of OS based on 100 randomly split testing sets was 0.78 (first, third quartiles = 0.77, 0.79).</p>Conclusions:<p>IL6 and HGF are potential predictive biomarkers of OS benefit from BEV + IFN in patients with mRCC. The model based on key biological and clinical factors demonstrated predictive efficacy for OS. These markers warrant further validation in future anti-VEGF and immunotherapy in mRCC trials.</p><p><i><a href="http://dx.doi.org/10.1158/1078-0432.CCR-22-0750" target="_blank">See related commentaries by Mishkin and Kohn, p. 2722</a> and <a href="http://dx.doi.org/10.1158/1078-0432.CCR-22-0027" target="_blank">George and Bertagnolli, p. 2725</a></i></p></div></jats:p>
Authors
Nixon, AB; Halabi, S; Liu, Y; Starr, MD; Brady, JC; Shterev, I; Luo, B; Hurwitz, HI; Febbo, PG; Rini, BI; Beltran, H; Small, EJ; Morris, MJ; George, DJ
MLA Citation
Nixon, Andrew B., et al. Data from Predictive Biomarkers of Overall Survival in Patients with Metastatic Renal Cell Carcinoma Treated with IFNα ± Bevacizumab: Results from CALGB 90206 (Alliance). 31 Mar. 2023. Crossref, doi:10.1158/1078-0432.c.6531354.
URI
https://scholars.duke.edu/individual/pub1571477
Source
crossref
Published Date
DOI
10.1158/1078-0432.c.6531354

Professor in Medicine
Contact:
133 Jones, Research Drive, Durham, NC 27710
Duke Box 2631, Durham, NC 27710