The GENETICS Company 

We are a dedicated team of entrepreneurs and scientists. Our focus is on the research, development and commercialization of products for the diagnosis, monitoring and therapy of patients suffering from neurodegenerative diseases and cancer. We have developed an impressive track-record of business and scientific achievements on our way to becoming an internationally competitive, self-sustainable and first class biopharmaceutical business.



Therapeutics Overview

The Company focuses on three programs, which aim at the identification, optimization and development of pioneering and innovative :

  • Small molecule Wnt-inhibitors for use in the treatment of colorectal cancer.
  • Small molecule BACE inhibitors against Alzheimer’s disease.
  • Small molecule inhibitors of cellular growth for use in cancer therapy.

Small Molecule Wnt-Inhibitors for Use in the Treatment of Colorectal Cancer

Colorectal cancer (CRC) remains one of the most common and aggressive malignancies globally. A critical pathway involved in CRC progression is the Wnt signaling pathway, which regulates essential processes like cell proliferation, differentiation, and apoptosis. Aberrant activation of this pathway is closely associated with tumorigenesis, particularly in CRC. In this context, small molecule Wnt-inhibitors have emerged as promising therapeutic agents for targeting this pathway and managing CRC. This article provides an overview of the molecular basis of the Wnt pathway in CRC and the potential of small molecule Wnt-inhibitors as a treatment option.

Mechanisms of Cellular Growth in Cancer

In normal cells, the growth and division of cells are tightly regulated through various signaling networks, ensuring that cell proliferation occurs only when necessary. This regulation is mediated by proteins such as cyclins, cyclin-dependent kinases (CDKs), and tumor suppressor genes, including p53. However, in cancer cells, these regulatory pathways are often disrupted, leading to unchecked cellular growth. Some common mechanisms contributing to uncontrolled growth in cancer include :

Small Molecule Wnt-Inhibitors: Mechanisms of Action

 Small molecule inhibitors are compounds that can modulate biological processes through their interaction with specific molecular targets. In the case of Wnt signaling, several small molecules have been identified that can disrupt different stages of the pathway. These inhibitors primarily target key proteins involved in the pathway, such as the Wnt ligands, Frizzled receptors, LRP5/6 co-receptors, or downstream β-catenin stabilization.

  • Inhibition of Wnt Ligand Binding : Some small molecules act by preventing the binding of Wnt ligands to Frizzled receptors. For instance, agents like LGK974 and OMP-54F28 function by targeting the Porcupine enzyme, which is involved in the secretion of Wnt ligands. By inhibiting this enzyme, the production of Wnt ligands is reduced, thus limiting their ability to activate the Wnt signaling pathway.
  • Disruption of Frizzled and LRP5/6 Receptor Interaction : Other small molecules focus on blocking the interaction between Frizzled receptors and LRP5/6 co-receptors, which is crucial for the initiation of Wnt signaling. Compounds such as ETC-159 have shown promise in inhibiting this interaction, thereby preventing the downstream activation of β-catenin.
  • Targeting β-Catenin Stabilization : Small molecules like ICG-001 and PRI-724 aim to inhibit the stabilization and nuclear translocation of β-catenin. These inhibitors function by disrupting the interaction between β-catenin and coactivators like CBP (CREB-binding protein), which are necessary for β-catenin-driven gene transcription.
  • Modulation of the Destruction Complex : The destruction complex, consisting of proteins like APC, AXIN, and GSK-3β, normally mediates the degradation of β-catenin. Some small molecules aim to restore the function of the destruction complex. For example, inhibitors of the casein kinase I (CKI) can enhance the activity of the APC/AXIN complex, leading to the degradation of β-catenin.

Potential of Small Molecule Wnt-Inhibitors in CRC Treatment

The therapeutic potential of small molecule Wnt-inhibitors in CRC is vast, as they can target the root cause of tumorigenesis in many CRC cases. Their ability to selectively inhibit specific components of the Wnt pathway offers several advantages over traditional chemotherapy, such as :

  • Targeted Therapy : Small molecule Wnt-inhibitors specifically target the Wnt signaling cascade, which is often dysregulated in CRC. This selective targeting reduces off-target effects and minimizes damage to healthy tissues.
  • Overcoming Resistance : Many CRCs develop resistance to conventional treatments such as 5-fluorouracil (5-FU) and oxaliplatin. Small molecules targeting the Wnt pathway may help overcome this resistance by addressing the underlying molecular drivers of tumorigenesis.
  • Combination Therapy : Small molecule Wnt-inhibitors may be combined with other treatment modalities, such as immune checkpoint inhibitors or chemotherapy, to enhance the overall therapeutic efficacy. The synergistic effects of combining different therapeutic approaches can help improve patient outcomes.
  • Personalized Medicine : The identification of specific mutations in the Wnt pathway could lead to personalized treatment strategies using small molecule inhibitors. Patients with CRC harboring mutations in APC, CTNNB1, or AXIN could benefit most from these targeted therapies

Small Molecule BACE Inhibitors Against Alzheimer's Disease

​Alzheimer's disease (AD) is a neurodegenerative disorder that leads to progressive cognitive decline and memory loss. It is characterized by the accumulation of amyloid-β (Aβ) plaques in the brain, which play a central role in the disease's pathophysiology. One of the key enzymes responsible for the generation of Aβ peptides is β-site amyloid precursor protein cleaving enzyme 1 (BACE1). Small molecule BACE inhibitors have gained significant attention as potential therapeutic agents for AD by targeting and inhibiting BACE1 activity, thus reducing the production of Aβ peptides. This article explores the role of BACE1 in Alzheimer's disease, the mechanism of action of BACE inhibitors, and their potential as a treatment option.

The Role of BACE1 in Alzheimer's Disease

The amyloid hypothesis of Alzheimer's disease posits that the accumulation of Aβ peptides in the brain leads to the formation of amyloid plaques, which subsequently trigger a cascade of neurotoxic events, including inflammation, oxidative stress, and neuronal damage. The production of Aβ peptides occurs through the cleavage of amyloid precursor protein (APP) by two enzymes: β-secretase (BACE1) and γ-secretase.

BACE1 is the first enzyme in the amyloidogenic processing pathway. It cleaves APP at the β-site, producing the soluble APPβ fragment and a membrane-bound C-terminal fragment (C99). The C99 fragment is then further cleaved by γ-secretase, releasing Aβ peptides of varying lengths, with Aβ42 being the most aggregation-prone and toxic form. The accumulation of Aβ42 in the brain is believed to be a primary driver of neurodegeneration in AD.

Given the central role of BACE1 in Aβ generation, inhibiting this enzyme represents a promising strategy for reducing amyloid plaque formation and slowing the progression of Alzheimer's disease.

Mechanism of Action of BACE Inhibitors

BACE inhibitors are small molecules that specifically target the active site of the BACE1 enzyme, preventing it from cleaving APP and producing Aβ peptides. These inhibitors work by binding to the catalytic site of BACE1, blocking its ability to recognize and process APP.

  • Binding to the Active Site : Most BACE inhibitors are designed to mimic the transition state of the substrate, allowing them to bind more tightly to the active site of the enzyme. This tight binding prevents BACE1 from interacting with its natural substrate, APP, thereby reducing the production of Aβ.
  • Selective Inhibition : The design of BACE inhibitors aims to ensure specificity for BACE1 over other enzymes that share similar catalytic sites, such as BACE2, which is involved in other physiological processes. This selectivity helps minimize off-target effects and adverse reactions.
  • Reduced Aβ Production : By inhibiting BACE1, the production of Aβ peptides is significantly reduced. This leads to a decrease in the formation of amyloid plaques, potentially preventing or slowing the neurotoxic effects associated with Aβ accumulation in Alzheimer's disease.

Types of Small Molecule BACE Inhibitors

Several types of small molecule BACE inhibitors have been developed, each with a different mechanism of action and chemical structure. These include :

  • Peptoid-Based Inhibitors : These inhibitors are designed to mimic the peptide substrate of BACE1, using peptoid structures to increase stability and bioavailability. They typically act by blocking the enzyme's active site, preventing APP cleavage.
  • Acyclic and Macrocyclic Compounds : Acyclic inhibitors are small, linear molecules that can penetrate the blood-brain barrier (BBB) and selectively inhibit BACE1. Macrocyclic inhibitors, on the other hand, are larger, ring-like molecules that offer increased binding affinity and selectivity for the BACE1 enzyme.
  • Transition-State Analogs : These inhibitors are designed to resemble the transition state of the BACE1-APP cleavage reaction. They mimic the structure of the intermediate complex formed during substrate processing, which allows for high specificity and potency.
  • Small Molecule Inhibitors with Allosteric Modulation : In addition to direct inhibition of the active site, some BACE inhibitors act by modulating the enzyme's allosteric sites. These compounds can regulate the enzyme's conformation, leading to a decrease in its activity without directly binding to the active site.

Clinical Development and Challenges

The development of BACE inhibitors as a therapeutic strategy for Alzheimer's disease has faced several challenges. While early preclinical studies demonstrated the potential of BACE inhibition to reduce Aβ levels and amyloid plaque formation, clinical trials have shown mixed results. Some of the main challenges include :

  • Blood-Brain Barrier (BBB) Penetration : For a BACE inhibitor to be effective in treating Alzheimer's disease, it must be able to cross the BBB, which protects the brain from potentially harmful substances. Achieving sufficient brain penetration is a key challenge for the development of small molecule inhibitors.
  • Efficacy in Humans : While BACE inhibition has shown promise in animal models, the results in human clinical trials have been less consistent. Some trials have failed to demonstrate significant cognitive improvements, despite reductions in Aβ levels. This suggests that Aβ may not be the sole driver of Alzheimer's disease, and additional mechanisms may need to be targeted for effective treatment.
  • Side Effects : Inhibiting BACE1 may have unintended con sequences due to its role in other physiological processes. For example, BACE1 is involved in the processing of other proteins, such as neuregulin-1, which is important for neuronal development and function. Inhibition of BACE1 could lead to side effects such as impaired synaptic plasticity, which may negatively impact cognitive function.
  • Long-Term Effects : Alzheimer's disease is a slow-progressing condition, and it may take years for the effects of BACE inhibition to become evident. Long-term studies are needed to assess the safety and efficacy of BACE inhibitors over extended periods.

Small Molecule Inhibitors of Cellular Growth for Use in Cancer Therapy

Cancer is a complex and heterogeneous disease characterized by uncontrolled cell growth, evasion of cell death, and metastasis. One of the primary hallmarks of cancer is the dysregulation of cellular growth and proliferation, which is often driven by abnormal signaling pathways. These pathways involve various growth factors, receptors, and intracellular signaling molecules that regulate cell division, survival, and metabolism. Small molecule inhibitors of cellular growth have emerged as an important class of therapeutic agents aimed at targeting the molecular mechanisms that drive cancer cell proliferation. This article explores the role of small molecule inhibitors in cancer therapy, their mechanisms of action, and their potential in clinical. applications.

Mechanisms of Cellular Growth in Cancer

 In normal cells, the growth and division of cells are tightly regulated through various signaling networks, ensuring that cell proliferation occurs only when necessary. This regulation is mediated by proteins such as cyclins, cyclin-dependent kinases (CDKs), and tumor suppressor genes, including p53. However, in cancer cells, these regulatory pathways are often disrupted, leading to unchecked cellular growth. Some common mechanisms contributing to uncontrolled growth in cancer include :

  • Activation of Oncogenes : Oncogenes, such as KRAS, MYC, and BCR-ABL, encode proteins that promote cell proliferation. Mutations or overexpression of these genes result in aberrant activation of signaling pathways that drive uncontrolled cell growth.
  • Dysregulation of Tumor Suppressor Genes : Tumor suppressor genes, such as TP53, PTEN, and RB, function to inhibit cell division and promote apoptosis. Loss of function of these genes can result in unregulated cell division and resistance to cell death signals.
  • Overactivation of Growth Factor Receptors : Growth factors like epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) bind to receptors on the cell surface, triggering downstream signaling that promotes cell division and survival. Overexpression or mutation of these receptors, such as in EGFR and HER2, leads to sustained activation of growth pathways.
  • Altered Cell Cycle Regulation : The cell cycle is controlled by a series of checkpoints that ensure proper progression through stages of cell division. Dysregulation of cell cycle checkpoints can result in uncontrolled proliferation, as seen in cancers with mutated CDK inhibitors like p21 or p27.

Role of Small Molecule Inhibitors in Cancer Therapy

Small molecule inhibitors target specific proteins and enzymes that are involved in the regulation of cell growth. These inhibitors have the advantage of being able to penetrate cell membranes and act intracellularly, allowing for more direct modulation of the growth signaling pathways. They can be used to inhibit specific molecular targets within cancer cells, interrupting key processes in cellular growth. The following are key targets of small molecule inhibitors in cancer therapy:

1. Inhibition of Tyrosine Kinase Receptors : Many growth factor receptors are receptor tyrosine kinases (RTKs), which play a central role in cellular signaling. Small molecules can inhibit the tyrosine kinase activity of these receptors, preventing the activation of downstream signaling pathways that promote cell growth. Notable examples include:

  • EGFR Inhibitors : Epidermal growth factor receptor (EGFR) is frequently overexpressed or mutated in several cancers, including non-small cell lung cancer (NSCLC) and colorectal cancer. Small molecule inhibitors like erlotinib and gefitinib block EGFR tyrosine kinase activity, thus preventing activation of the PI3K/Akt and Ras/MAPK signaling pathways that drive cancer cell proliferation.
  • HER2 Inhibitors : HER2 is overexpressed in certain breast cancers, leading to aggressive growth. Trastuzumab (Herceptin) and lapatinib are small molecules targeting HER2, inhibiting its kinase activity and impeding tumor growth.
  • VEGFR Inhibitors : Vascular endothelial growth factor receptor (VEGFR) is critical for angiogenesis, the process of new blood vessel formation that supplies nutrients to tumors. Small molecules such as sorafenib and sunitinib inhibit VEGFR activity, thereby limiting tumor growth and metastasis by starving the tumor of nutrients.

2. CDK Inhibitors : Cyclin-dependent kinases (CDKs) are central regulators of the cell cycle, promoting progression through the G1, S, G2, and M phases. Aberrant activation of CDKs, particularly CDK4/6, contributes to uncontrolled cell division in many cancers. Small molecule inhibitors, such as palbociclib, ribociclib, and abemaciclib, specifically inhibit CDK4/6, leading to cell cycle arrest in the G1 phase and preventing cancer cell proliferation.

3. Inhibition of PI3K/Akt/mTOR Pathway : The PI3K/Akt/mTOR signaling pathway plays a critical role in promoting cell survival, growth, and metabolism. Dysregulation of this pathway is common in various cancers. Small molecules targeting components of this pathway, such as:

  • PI3K Inhibitors : Drugs like idelalisib target PI3K, inhibiting its activation and subsequent Akt/mTOR signaling.
  • Akt Inhibitors : Small molecules like ipatasertib directly target Akt, preventing its activation and impeding cell survival and proliferation.
  • mTOR Inhibitors : Temsirolimus and everolimus inhibit mTOR, a key downstream target in the PI3K/Akt pathway, thus reducing tumor cell growth and promoting cell death.

4. Histone Deacetylase (HDAC) Inhibitors : HDACs are enzymes that regulate gene expression by modifying histone proteins, leading to chromatin condensation and suppression of gene transcription. In cancer, dysregulation of HDAC activity can result in the silencing of tumor suppressor genes. HDAC inhibitors like vorinostat and panobinostat can restore the expression of tumor suppressor genes and induce apoptosis in cancer cells.

5. Proteasome Inhibitors : The proteasome is responsible for degrading unwanted or damaged proteins in the cell. In cancer, the proteasome is often overactive, leading to the degradation of pro-apoptotic proteins and promotion of tumor growth. Bortezomib and carfilzomib are proteasome inhibitors that block protein degradation, leading to the accumulation of damaged proteins and the induction of cancer cell death.

6. Apoptosis Induction : Apoptosis is a form of programmed cell death that is often defective in cancer cells. Small molecule inhibitors can activate pro-apoptotic pathways in cancer cells, triggering cell death. For example, small molecules targeting the BCL-2 family of proteins, such as venetoclax, can promote apoptosis in hematologic malignancies like chronic lymphocytic leukemia (CLL). 

Clinical Applications and Challenges

 While small molecule inhibitors have shown significant promise in cancer therapy, several challenges remain :

  • Dru g Resistance : Cancer cells can develop resistance to small molecule inhibitors through various mechanisms, including mutation of the target protein, activation of alternative pathways, and changes in drug metabolism. Overcoming resistance remains a critical challenge in cancer therapy.
  • Toxicity and Side Effects : While small molecule inhibitors are designed to specifically target cancer cells, they may also affect normal cells that share similar signaling pathways. This can result in off-target toxicity and side effects such as gastrointestinal issues, fatigue, and cardiovascular toxicity.

Combination Therapies : To improve efficacy and overcome resistance, small molecule inhibitors are often used in combination with other therapies, such as chemotherapy, immunotherapy, or targeted therapies. Combination treatments hold great promise for enhancing treatment outcomes and reducing the likelihood of resistance.

  • Personalized Medicine : The heterogeneity of cancer means that not all patients will respond to a given small molecule inhibitor. Personalized medicine, which tailors treatment based on the genetic and molecular profile of a patient's tumor, is essential for optimizing therapy and improving patient outcomes.