Your analysis
Multidimensional precision diagnostics form the basis for targeted therapy concepts.

The Hallwang Clinic combines evidence-based medicine with the latest diagnostic technologies to provide patients from all over the world with advanced or metastatic cancer access to the highest diagnostic precision and the most individualized therapeutic approaches.
Our mission is to provide diagnostics at the level of leading international cancer centers — enhanced by the innovative strength, speed, and flexibility of an independent, highly specialized private clinic.
We are rethinking diagnostics: integrative, multidimensional, and without the limitations of traditional standard approaches. Our goal is to prepare the best possible, scientifically sound treatment decision for each individual patient.
Why diagnostic quality determines therapeutic success
Stage 4 cancer and metastatic tumor diseases in particular are highly complex biologically, with different subclones, resistance mechanisms, changes in the microenvironment, and a dynamic disease progression. Conventional diagnostics often do not adequately reflect this diversity.
That is why we rely on multidimensional precision diagnostics that holistically analyze the tumor, immune system, and microenvironment. Only with this depth of diagnostics can targeted therapies, modern immunotherapies, innovative off-label treatments, and personalized combinations be planned sensibly and safely.
Our diagnostic philosophy follows three principles:
As comprehensive as necessary: We think of diagnostics without limitations imposed by rigid standard paths.
As precise as possible: We use state-of-the-art genome, immune, and liquid biopsy technologies.
As individual as the tumor itself: We rely on personalized analysis instead of a “one size fits all” approach.
Our approach to precision diagnostics
Our approach is based on a clearly structured, state-of-the-art multi-omics framework that covers all relevant levels of tumor and immune biology. Each layer of analysis provides crucial information for treatment decisions in metastatic diseases.
We integrate three key diagnostic areas:
1. Molecular tumor diagnostics (tissue-based genomics)
Molecular analysis of the tumor is the indispensable foundation of modern precision oncology. Using state-of-the-art sequencing technologies and bioinformatic platforms, we create a complete, high-resolution molecular profile to identify optimal, personalized therapeutic approaches.
I. Comprehensive tumor profiling: DNA and RNA
These methods are used to maximally capture genetic and transcriptional changes in order to identify therapeutic targets and resistance mechanisms.
- Next-generation sequencing panels (NGS): These panels specifically examine a large number of cancer-relevant genes in a single test run and identify mutations, fusions, and structural changes that represent therapeutic targets. They form the basis of many targeted therapies and modern immune strategies.
- Whole-exome sequencing (WES): WES analyzes all protein-coding regions of the genome where the majority of disease-relevant mutations occur. This allows the detection of rare or complex tumor drivers that would be overlooked in standard panels.
- Whole-genome sequencing (WGS): WGS examines the entire genome, including non-coding regions and structural variants. This enables a particularly detailed classification of tumors with unusual genetics or mixed patterns.
- RNA-Seq (transcriptome sequencing): RNA-Seq captures gene expression patterns and active signaling pathways in the tumor, revealing subtypes, resistance mechanisms, or immunological activity profiles. This data helps to select targeted therapies even more precisely.
- Fusion Gene Detection (NGS-based / FISH): Therapy-relevant gene fusions such as ALK, ROS1, or NTRK are reliably detected using NGS or FISH. These fusions are highly specific targets for approved and experimental targeted therapies.
- Copy-Number Variation (CNV) Analysis: CNV analyses identify amplifications or deletions that can confer growth advantages. These markers are important for prognosis, therapy decisions, and drug selection, for example in the case of HER2 or MET amplifications.
- FISH (Fluorescence In Situ Hybridization): FISH allows visual confirmation of gene amplifications or rearrangements directly in tumor tissue. The procedure is particularly valuable when NGS results need to be verified or unclear findings need to be clarified.
- DNA methylation profiling: Analysis of epigenetic patterns allows tumors to be classified more precisely and subtypes to be identified that are morphologically difficult to distinguish. Methylation profiles are increasingly being used for complex or rare tumors.
II. Predictive markers for immune and DNA-damaging therapies
The analysis of specific repair and stability markers is crucial for selecting the optimal systemic therapy.
- DNA repair gene analysis (BRCA1/2, PALB2, ATM, CHEK2, etc.): Defects in DNA repair genes lead to specific vulnerabilities in the tumor that can be exploited therapeutically — for example, with PARP inhibitors or platinum-based chemotherapies. The analysis of these genes is a central component of modern precision medicine.
- HRD testing (homologous recombination deficiency): HRD indicates whether homologous recombination is impaired – a strong predictive marker for response to DNA-damaging therapies. Tumors with HRD often have a particularly high mutation rate and immunological attack points.
- Mismatch repair, MSI-H, and tumor mutation burden (TMB): Tumors with MSI-H or high TMB carry many mutations and neoantigens, which is why they respond particularly well to immune checkpoint inhibitors. These markers are now established and indispensable parameters for immunotherapy planning.
III. Pathology and tumor microenvironment (TME) analysis
These tissue-based analyses provide functional and spatial information about tumor activity, immune infiltration, and the cellular environment.
- IHC Advanced Panels: Using immunohistochemical staining, we determine markers such as PD-L1 or Ki-67, which allow conclusions to be drawn about tumor activity, cell division, and potential immunotherapy targets. IHC is a central component of any tumor classification.
- Digital pathology and AI-assisted analytics: With the help of artificial intelligence, we analyze digital tissue images, recognize microstructures, and quantify cell populations more accurately than with purely manual methods. This significantly improves diagnostic precision and reproducibility.
- Multiplex IHC / Multiplex IF: These methods allow the simultaneous detection of many immune markers in a single tissue section. This enables us to precisely map complex interactions between tumor cells, immune cells, and stromal components.
- Spatial Transcriptomics: Spatial transcriptomics combines tissue imaging and gene expression to reveal which genes are active in which parts of the tumor. This provides profound insights into heterogeneity, immune infiltration, and resistance clusters.
- Proteomics: Proteomics analyzes thousands of proteins simultaneously and reveals functional changes in tumor metabolism, signaling pathways, or stress response. The method complements genomic testing with a function-oriented level.
This comprehensive molecular profile lays the foundation for targeted, immunological, and innovative personalized therapy approaches for each individual patient.
2. Blood-based diagnostics (liquid biopsy and immune profiling)
Metastatic tumors are dynamic and develop resistance during therapy. Liquid biopsy enables non-invasive real-time analysis of tumor biology — a decisive advance in precision oncology for advanced stages of disease. It is particularly valuable when traditional tissue biopsies are risky or difficult to perform.
I. Liquid biopsy: Tumor DNA and cells
These procedures analyze circulating material from the tumor in the blood to monitor current genetic changes with high sensitivity.
- ctDNA analyses: Circulating tumor DNA allows highly sensitive detection of current mutations and resistance mechanisms – often earlier than imaging techniques. ctDNA enables MRD monitoring, real-time therapy adjustments, and early detection of relapse.
- CTC analysis: Circulating tumor cells provide access to living tumor cells that can be characterized directly. They provide insights into metastatic mechanisms, tumor heterogeneity, and parallel subclone developments.
- Multi-analyte liquid biopsy: By simultaneously analyzing ctDNA, CTCs, RNA, proteins, and exosomes, we gain a particularly deep and dynamic picture of tumor biology. This method often detects changes earlier than any other diagnostic technique.
- Digital Droplet PCR (ddPCR): ddPCR detects individual mutation events with extremely high sensitivity and is ideal for MRD detection or close monitoring during therapy.
II. Advanced fluid and immune profiles
These profiles shed light on the interaction between the tumor and the immune system, which is essential for planning immune and cell therapies.
- Immune cell and HLA profiling: We record the composition and activity of the immune system and analyze HLA characteristics that may be crucial for immunotherapies and personalized vaccination strategies.
- Cytokine and chemokine profiling: These profiles provide insight into inflammatory processes and immune activity, which is important for assessing immune fitness and potential immunotherapeutic targets.
- Exosome analysis and cfRNA: Exosomes and cell-free RNA carry signatures of tumor development and enable diagnostics even when ctDNA is low. They open up new insights into the development of resistance and metastasis.
- Tumor-Educated Platelets (TEP): Tumor-induced changes in platelet RNA can provide early indications of tumor activity — a new, promising biomarker.
- TCR/BCR sequencing: By analyzing the T and B cell repertoire, we can map the body’s immune response in detail and identify which immune strategies might be most appropriate.
Liquid biopsy is particularly valuable for patients for whom conventional biopsies are risky or difficult to perform.
3. Functional and integrated diagnostics
Beyond pure genetic analysis, we functionally evaluate how the tumor responds to therapeutic agents. This level of personalized precision is crucial for modeling and validating the actual effectiveness of treatments in advance.
I. Functional and ex vivo testing
These methods use living tumor material to test the effectiveness of drugs under realistic conditions outside the body.
- Advanced chemosensitivity analyses: We test the patient’s own tumor cells ex vivo against a variety of drugs to predict sensitivity or resistance. This is particularly valuable in the case of multiple pretreated or rare tumors.
- Functional tests from blood or tissue: CTCs or tumor biopsies are cultured and examined for their response to drug classes. This provides us with real functional patterns and allows us to prioritize specific therapeutic approaches.
- Patient-derived organoids (PDOs): 3D mini-organoids replicate the patient’s tumor almost identically. They are ideal for testing drugs and combinations under realistic conditions.
- Patient-derived xenografts (PDX): Upon request, tumor cells can be tested in mouse models to examine long-term drug profiles — an option for particularly complex cases.
- Metabolomics: This analysis records metabolic changes in the tumor and identifies metabolic weaknesses that can be exploited therapeutically.
II. Advanced imaging and AI analysis
Modern imaging and artificial intelligence (AI) extract hidden information about tumor aggressiveness and response probability in a non-invasive manner.
- FAPI-PET: FAPI-PET detects fibroblastic activity in the tumor – often more sensitively than classic FDG-PET – and is particularly helpful in tumors with low glucose uptake.
- Radiomics: AI-based analysis extracts quantifiable patterns from CT, MRI, or PET data and detects changes that are barely visible to the human eye.
- AI-assisted image analysis: Neural networks support prognosis modeling, classification, and therapy planning based on large image datasets.
III. Integral multiomics and AI modeling
The consolidation of all data in advanced computational models enables the most accurate prediction of treatment success.
- Multiomics integration: We integrate genomics, proteomics, metabolomics, and immune profiles to generate a complete picture of tumor biology.
- AI-driven therapy prediction models: Modern algorithms model the response probabilities of different therapies, prioritize active substances, and identify promising combinations.
These functional and computer-assisted analyses enable us to refine therapy decisions scientifically — especially when several treatment strategies are possible or established options have been exhausted.
For whom is our diagnostic program particularly suitable?
Our precision diagnostics are particularly aimed at people with metastatic disease or stage 4 cancer who, despite intensive prior treatment, have not received a clear therapy recommendation or whose tumor has already developed resistance patterns.
It is also suitable for patients who seek a second, independent opinion or want targeted access to innovative, personalized treatment strategies.
Especially in cases of complex disease progression or when several treatment options are available, maximum diagnostic precision provides the basis for individualized, well-founded, and new therapeutic perspectives.
Expertise as the key to true precision oncology
Such a comprehensive, multidimensional diagnostic approach is not standard worldwide — neither in routine care nor in many specialized centers. But it is precisely this depth that is required to initiate truly precision-based therapy that understands the tumor in all its genetic, immunological, and functional facets.
In order to interpret these diagnostics meaningfully and implement them therapeutically, a highly specialized, multidisciplinary team of oncologists is needed that is familiar with the possibilities of modern molecular medicine, immuno-oncology, and functional diagnostics. Precision medicine is not just a question of technology — it requires experienced experts who can clinically classify genetic patterns, immunological signatures, resistance mechanisms, and complex tumor interactions correctly.
The Hallwang Clinic combines this expertise in a team of board-certified oncologists and specialized diagnostic partners. In addition, we are closely integrated into an international network of university research centers, laboratory partners, specialist clinics, and translational science teams that continuously translate cutting-edge research into clinically useful strategies.
This ecosystem enables us not only to perform even the most advanced diagnostics, but also to make them medically useful and applicable to individual patients.
Your access to world-leading precision diagnostics
Contact us for an individual case assessment or send us your medical records for a precise second opinion. We support you with state-of-the-art diagnostics, a specialized team of experts, and a clear goal: to reveal new options.