You are currently viewing a new version of our website. To view the old version click .
  • Indexed in
    Scopus
  • 24 days
    Time to First Decision

Onco

Onco is an international, peer-reviewed, open access journal on the whole field of oncotargets and cancer therapies research published quarterly online by MDPI.

All Articles (137)

Pre-Existing Immunity Shapes Cancer Immunotherapy Efficacy

  • Anastasia Xagara,
  • Filippos Koinis and
  • Konstantinos Tsapakidis
  • + 6 authors

Immunotherapy has revolutionized the management of patients with cancer. Immune checkpoint inhibition (ICI) is a promising treatment option that targets the molecular mechanisms that cancer cells exploit to prevent immune-mediated elimination. ICI therapy can cause exceptional long-term tumor remissions, in some cases, even after treatment discontinuation. Despite its success, many patients acquire resistance or fail to respond due to immune escape mechanisms mediated by the tumor and its microenvironment. Pre-existing immunity status of individuals seems to play a fundamental role in immunotherapy response and eventually tumor progression, as it orchestrates tumor-immune interactions. Different immune cell subsets, both in the tumor microenvironment and the peripheral blood, are established mediators that contribute to immune escape in various tumor types. Based on these findings, the elucidation of the mechanisms implicated in the regulation of these immune cells has become a priority for investigators focused on improving the efficacy of ICI. This will be essential for identifying responders as well as for developing novel therapeutic modalities to improve clinical outcomes. Herein, we summarize preclinical and clinical evidence proposing a predictive role of pre-existing immunity for clinical responses to immunotherapies.

7 January 2026

ICI efficacy depends on the composition of immune cells in TME. (a) Immune excluded tumors without pre-existing immune T cells do not respond to ICI immunotherapy. They are characterized by impaired T-cell infiltration and low PD-L1 expression. (b) Immune suppressive tumors contain a high density of immunosuppressive cells, such as Tregs and MDSCs. (c) Immune infiltrated tumors respond better to immunotherapy due to the presence of pre-existing immune T cells, high levels of PD-L1, and exhausted T cells.

Background: There are approximately 5.4 M basal cell (BCC) and squamous cell (SCC) carcinomas diagnosed each year, and the number is increasing. Currently, the gold standard for skin cancer diagnosis is histopathology, which requires the surgical excision of the tumor followed by pathological evaluation of a tissue biopsy. The three-dimensional (3D) nature of human tissue suggests that two-dimensional (2D) cross sections may be insufficient in some cases to represent the complex structure due to sampling bias. There is a need for new techniques that can be used to classify skin lesion types and margins noninvasively. Methods: We use optical coherence tomography volume scan images and AI to noninvasively create 3D images of basal cell and squamous cell carcinomas. Results: Three-dimensional optical coherence tomography images can be broken down into a series of cross sections that can be classified as benign or cancerous using convolutional neural network models developed in this study. These models can identify cancerous regions as well as clear edges. Cancerous regions can also be verified based on visual review of the color-coded images and the loss of the green and blue subchannel pixel intensities. Conclusions: Three-dimensional optical coherence tomography cross sections of cancerous lesions can be collected noninvasively, and AI can be used to classify skin lesions and detect clear lesion edges. These images may provide a means to speed up treatment and promote better patient screening, especially in older patients who will likely develop several lesions as they age.

5 January 2026

Three-dimensional OCT images of a basal cell carcinoma (BCC) (A) and normal skin (B) obtained in vivo using OptoScope. Three-dimensional reconstructions of a BCC (A) and normal skin (B) made using the volume scan app on the OptoScope. The 3D reconstruction can be cut at any point as demonstrated in Figure 2 to identify where the lesion is located. The 3D reconstructions were created by combining 128 horizontal scans using MATLAB R2024B. The lesion is predicted to be a BCC based on the CNN model and the evaluation of slice #64 of the cross section shown in Figure 2 (see Table 1). The yellow color is that of the stratum corneum and the blue represents the collagen of the papillary dermis.

Tumor Microenvironment: Current Understanding and Therapeutic Implications in Adult and Pediatric Cancers

  • Satyendra Batra,
  • Prashant Prabhakar and
  • Debabrata Mohapatra
  • + 5 authors

The tumor microenvironment (TME) plays an important role in the development, progression, and treatment response of pediatric cancers, yet remains less elucidated compared to adult malignancies. Pediatric tumors are unique with a low mutational burden, an immature immune landscape, and unique stromal interactions. The resultant “cold” immune microenvironments limits the effectiveness of conventional immunotherapies. This review summarizes the key cellular and non-cellular components of the pediatric TME—including T cells, NK cells, tumor-associated macrophages, cancer-associated fibroblasts, extracellular matrix remodeling, angiogenesis, and hypoxia—and describes how these elements shape tumor behavior and therapy resistance. The role of TME in common pediatric cancers like leukemia, lymphoma, neuroblastoma, brain tumors, renal tumors, and sarcomas is discussed. Emerging therapeutic strategies targeting immune checkpoints, macrophage polarization, angiogenic pathways, and stromal barriers are discussed.

25 December 2025

Role of M1 and M2 macrophages in tumor progression. M1macrophages secrete pro-inflammatory cytokines, leading to their anti-tumor effect, while M2 macrophages have anti-inflammatory activity leading to tumor formation and progression. Created in BioRender. Batra, S. (2025) https://BioRender.com/ija1wc4 (accessed on 18 October 2025).

Background: Melanoma and triple-negative breast cancer (TNBC) are the most aggressive skin and breast cancers, often diagnosed at late stages with limited treatment options. The melanoma-associated antigen melanotransferrin (MTf) is overexpressed in these solid tumors, where it drives tumorigenesis, progression, and chemoresistance. Its inhibition correlates with tumor regression, making MTf a promising therapeutic target. This study aimed to develop a novel, selectively targeted antibody–drug conjugate (ADC) against MTf-expressing melanoma and TNBC cancer cells using SNAP-tag fusion protein conjugation technology. Methods: We generated an L49(scFv)-SNAP-tag antibody fusion protein engineered through the genetic fusion of a humanized anti-MTf single-chain variable fragment (scFv) with a SNAP-tag fusion protein capable of site-specific self-labelling with O6-benzylguanine (BG) modified substrates in 1:1 stoichiometry. Binding and internalization of the conjugate labeled with BG-Alexa 488 (L49(scFv)-SNAP-Alexa488) were assessed by confocal microscopy and flow cytometry in MTf-overexpressing cell lines. Cytotoxicity was evaluated using the cell viability XTT assay after conjugating the SNAP-fusion protein to the potent monomethyl auristatin-F (BG-AURIF). Results: The L49(scFv)-SNAP-Alexa488 conjugate demonstrated specific binding and internalization into MTf-positive melanoma and TNBC cells. The corresponding ADC, L49(scFv)-SNAP-Linker-AURIF, exerted potent, antigen and dose-dependent cytotoxicity, with IC50 values in the nanomolar range (4.77–34.43 nM). Conclusions: We successfully generated a novel SNAP-tag-based ADC that selectively eliminates MTf-overexpressing tumor cells. This proof-of-concept highlights MTF’s value as a therapeutic target and demonstrates that a smaller-format, non-cleavable linker SNAP-tag-based ADC can achieve potent nanomolar cytotoxicity, supporting further development of MTF-targeted immunotherapies for melanoma and TNBC.

25 December 2025

Molecular cloning and protein characterization of L49(scFv)-SNAP: (a) Schematic representation of the open reading frame (ORF) of pCB-L49(scFv)-SNAP mammalian expression plasmid. (b) Microscopy visualization of transfected cells after enrichment of eGFP-positive cells to more than 90% using Zeocin treatment. Pictures show the bright and green channels of the same field, captured using a ZOE™ Fluorescent Cell Imager (100 µm scale). (c) Elution profile of L49(scFv)-SNAP by IMAC using the ÄKTA Avant 25 system. The x-axis represents the flow-through volume over time, and the y-axis indicates absorbance in milliabsorbance units (mAU). The green line indicates the imidazole gradient applied to successfully elute L49(scFv)-SNAP within a single peak (filled blue curve). (d) Characterization of eluted protein. Concentrated protein was run on two 10% SDS-PAGE gels alongside a protein standard (line 1). The first gel was stained using AcquaStain solution (line 2), and the second was transferred to a PVDF blotting membrane and incubated with mouse Anti-polyHistidine−HorsePeroxidase antibody and revealed using a blotting buffer (line 3). L49(scFv)-SNAP was successfully conjugated to BG-Alexa 488 fluorophore as visualized using a Dark Reader Transilluminator (line 4). Blue arrows indicate the presence of L49(scFv)-SNAP at the expected molecular weight. Sfi1 and Not1: restriction enzyme sites; scFv: single-chain variable fragment; eGFP: enhanced green fluorescent protein; kDa: kilodalton; MW: molecular weight.

News & Conferences

Issues

Open for Submission

Editor's Choice

Reprints of Collections

Artificial Intelligence in Cancer, Biology and Oncology
Reprint

Artificial Intelligence in Cancer, Biology and Oncology

Editors: Hamid Khayyam, Ali Hekmatnia, Rahele Kafieh
Soft Tissue Sarcomas: Treatment and Management
Reprint

Soft Tissue Sarcomas: Treatment and Management

Editors: Shinji Miwa, Po-Kuei Wu, Hiroyuki Tsuchiya

Get Alerted

Add your email address to receive forthcoming issues of this journal.

XFacebookLinkedIn
Onco - ISSN 2673-7523