Translational Oncology

Our research aims to respond to the clinical needs of oncology patients by identifying predictive markers of response and new therapeutic combinations. DR. RUBEN PIO AND DR. ANTONIO GONZÁLEZ
PRINCIPAL INVESTIGATORS OF THE TRANSLATIONAL ONCOLOGY GROUP

The Translational Oncology Research Group of the Cima Solid Tumor Program focuses primarily on two tumor types: ovarian cancer and lung cancer.

Ovarian cancer is a leading cause of death among women and the most lethal cause of death from gynecologic malignancy. Traditional management of ovarian cancer (surgery and chemotherapy) allows temporary disease control, but most treated patients will relapse. Maintenance therapy based on a new generation of PARP inhibitors (PARPi) has prolonged the time during which the disease does not progress. However, this new approach is not yet sufficient to keep the disease under control.

New combinatorial strategies for treating recurrent ovarian cancer are currently being investigated, and immunotherapy is one of the most promising.

In the case of lung cancer, the type of tumor that causes the most deaths in the world, immunotherapy is already a clinical reality. Despite this, the percentage of patients who benefit from this type of therapy, mainly based on inhibition of the PD-1/PD-L1 immune checkpoint, is around 30%.

Therefore, for the best clinical management of patients with ovarian or lung cancer, it is necessary to optimize current therapeutic strategies and rationalize their use by applying predictive markers of response or resistance.

In our group, we investigate the identification of these biomarkers and develop new therapeutic strategies based on the potentiation of the antitumor immune response by modulation of the complement system, both in ovarian and lung cancer.

We have a multidisciplinary team supported by close collaboration with other national and international groups, as well as sophisticated animal models and cutting-edge technologies for identifying biomarkers, such as multispectral immunophenotyping or multiple protein quantification by Luminex.

GROUP LEADERS

Dr. Rubén Pío
+34 948 194 700 | Ext. 812020
rpio@unav.es
Research profile
   
Dr. Antonio González
+34 948 194 700
agonzalezma@unav.es
Research profile

Objectives of the Translational Oncology Group

We do research to improve the treatment, prognosis and prediction of response to treatment of patients with lung or ovarian cancer.

Identifying biomarkers
Characterize vulnerabilities in tumors resistant to targeted therapy or immunotherapy to identify predictive markers of response or resistance.

Modulate the complement system
To develop new therapeutic combinations that reverse resistance to targeted therapies or immunotherapies, mainly based on the modulation of the complement system.

Design new therapeutic strategies
To develop new therapeutic strategies with immunostimulatory activity in lung and ovarian cancer models. 

IDENTIFYING VULNERABILITIES

More effective therapies for lung tumors

Lung tumors with co-mutations in the KRAS oncogene and the tumor suppressor gene LKB1/STK1 show high resistance to immunotherapy based on PD-1/PD-L1 inhibition. In collaboration with the Department of Anatomic Pathology of the Clínica Universidad de Navarra, the pharmaceutical industry, and other national and international research groups, we are carrying out a detailed characterization of this type of tumor at genomic, transcriptomic, and phenotypic levels (in the image). The objective is to identify vulnerabilities that will allow the development of more effective therapies for this type of tumor.

Lines of research

PIs: Antonio González and Beatriz Tavira

Ovarian cancer is one of the leading causes of death among women and the most lethal cause of death from gynecologic malignancy. Traditional management of ovarian cancer (cytoreductive surgery and platinum-based chemotherapy) allows for temporary disease control, but 70% of treated patients relapse within the first three years.

Maintenance therapy based on a new generation of poly (ADP-ribose) polymerase inhibitors (PARP inhibitors or PARPi) has prolonged the time during which the disease does not progress. However, this new approach is not yet sufficient to keep the condition under control. Therefore, new combinatorial strategies for treating recurrent ovarian cancer are currently being investigated, and immunotherapy is one of the most promising. 

Several ongoing clinical trials are studying the effect of immunotherapy based on PD-1/PD-L1 inhibition and PARPi as part of maintenance therapy. Some of these trials are being led by researchers at the Clínica Universidad de Navarra and represent an opportunity for improving patients and identifying biomarkers associated with response or resistance to these new treatments. Specifically, the Spanish Ovarian Cancer Research Group (GEICO) has launched the phase III clinical trial ANITA (Atezolizumab and Niraparib Treatment Association), led by Dr. Antonio González-Martín, clinical PI of the Translational Oncology group. This study, which collaborates with several gynecological cancer research groups in Europe through the ENGOT network, includes patients with ovarian cancer who have previously received platinum treatment and have relapsed more than six months after the last treatment. These patients receive a platinum-based regimen followed by a PARPi (Niraparib) and are randomized to receive a PD-L1 inhibitor (atezolizumab) for the duration of treatment. 

This study's main objective is to identify predictive biomarkers of response/resistance to these drugs (PARPi and anti-PD-L1) and to create response profiles. This, would allow early selection of each patient based on their biological characteristics (genetic and immunological) to which specific treatment would be assigned according to their probability of response, allowing personalized treatment to be obtained. This personalized medicine approach opens the door to the selection from the outset of those women who are likely to respond, avoiding the appearance of unnecessary side effects due to the inappropriate use of drugs, improving the quality of life of patients, and saving costs for the healthcare system.

On the other hand, another of the present lines focuses on the study of changes in the tumor microenvironment induced by neoadjuvant chemotherapy with or without anti-angiogenic drugs (bevacizumab) in patients with recurrent ovarian cancer (MINOVA study, GEICO 1205).

In both projects, biological material (tumor tissue, blood, serum, or plasma) will be used to identify biomarkers associated with predicting response or resistance by analyzing changes in the tumor microenvironment after administration of the therapies.

In parallel, animal models of ovarian cancer will also be used to gain a more detailed understanding of the complexity of the biological pathways involved in the origin and development of this disease, as well as the mechanisms of metastasis and action of resistance to the different treatments used in the management of the disease. In this aspect, one of the main avenues of current study focuses on the analysis of the effect on the tumor microenvironment and metabolism of the combination of caloric restriction drugs and current ovarian cancer management treatments such as PARPi and immunotherapy. 

General objectives:

  • Identifying biomarkers associated with response/resistance to the combination of PARP inhibitors and immunotherapy by high-throughput analysis techniques (proteomics by mass spectrometry and multiple protein quantification by Luminex) in plasma. 
  • Characterization of changes in the tumor microenvironment by multispectral immunophenotyping (Vectra Polaris) in patients with recurrent ovarian cancer after administration of different neoadjuvant therapies.
  • Generation of preclinical models of ovarian cancer and their application for the study of therapies and models of resistance and metastasis.

PI: Rubén Pío

The efficacy of antitumor immunotherapy is limited by intrinsic genetic factors. Mutations in KRAS, STK11 or TP53 define distinct lung tumor subgroups with clear biological and therapeutic differences. Tumors with KRAS/STK11 mutations express lower levels of immune activation markers, including PD-L1, and show a higher number of neutrophils, possibly with immunosuppressive capacity. In contrast, tumors with KRAS/TP53 mutations show a higher mutational burden and denser T-cell and NK-cell infiltration. Consistent with these observations, clinical response rates differ significantly between tumors with STK11 or TP53 mutations. Therefore, new therapeutic strategies that reverse the resistance of tumors with an immunosuppressive environment, such as STK11 mutated tumors, are required. 

Objectives

  • Generation of a repository of KRAS/STK11mut and KRAS/TP53mut human lung adenocarcinoma samples.
  • Immunological characterization of KRAS/STK11mut and KRAS/TP53mut human lung adenocarcinomas.
  • Generation and characterization of isogenic mouse models of KRAS/STK11mut and KRAS/P53mut lung adenocarcinomas
  • Preclinical evaluation of novel IO combinations for KRAS/STK11mut lung adenocarcinomas

This multicenter project, which also involves the Vall d'Hebon Cancer Institute and the PeterMac Cancer Center, will provide new insights into the oncogenic immunoediting mechanisms that drive primary resistance to PD-1 axis agents and will enable the development of new therapeutic strategies to overcome this resistance.

PIs: Rubén Pío and Daniel Ajona

The efficacy of antitumor immunotherapy is limited both by factors intrinsic to the tumors and by the presence of an immunosuppressive tumor microenvironment. In addition, a significant number of patients suffer from adverse effects related to immunotherapy.

Our group has demonstrated, in preclinical models, that inhibition of the complement C5a/C5aR1 pathway reverses the resistance of non-small cell lung tumors (NSCLC) to PD-1/PD-L1 blockade. This finding has laid the groundwork for a clinical trial evaluating combined C5aR1 and PD-L1 inhibition in solid tumors (STELLAR-001; NCT03665129). Moreover, several groups have demonstrated that C5a/C5aR1 inhibition in mice attenuates colitis, one of the most frequent adverse effects of immunotherapy.

In the present project, we aim to exploit the potential of complement modulation, in particular of C5a/C5aR1, to reverse resistance to PD-1/PD-L1 blockade while attenuating its adverse effects. We also want to apply our recent discovery of the use of fasting as an enhancer of the effect of PD-1 blockade.

Objectives:

  • Characterization of the function of complement proteins in the biology of cell subpopulations in the tumor microenvironment.
  • Identification of complement-related traits and markers associated with immunotherapy resistance.
  • Development of new drugs targeting the C5a/C5aR1 axis
  • Evaluation of new immunotherapy combinations based on our findings.

Meet the research team

Scientific activity of the
Translational Oncology Research Group