top of page
  • What is the regulatory mechanism of a kinase?
    Kinases are types of enzymes that add a phosphate group to other molecules – a process called phosphorylation. This is a crucial process in regulating the function of proteins in our cells, affecting their activity, stability, and location. Here's a simple way to understand the regulatory mechanism of a kinase: Activation: Before a kinase can do its job, it often needs to be activated. This is sometimes achieved by the addition of a phosphate group to the kinase itself by another kinase. Substrate Binding: Once the kinase is activated, it needs to find the correct target protein, known as a substrate. The kinase has a specific area, called a binding site, that fits like a puzzle piece with its substrate. Only certain substrates can fit into this binding site, which ensures the kinase modifies the correct proteins. Phosphorylation: After the substrate is bound to the kinase, the kinase adds a phosphate group to the substrate. The addition of the phosphate often changes the shape of the substrate, which can activate or deactivate the substrate protein, or alter its location or interaction with other proteins. Deactivation: Some kinases are self-regulating and can deactivate themselves by adding phosphate groups to their own structure. Other times, separate enzymes, called phosphatases, remove the phosphate group from the kinase, deactivating it. This system is a significant part of how cells control their internal processes. For instance, when a hormone arrives at a cell, it can cause a series of activations of kinases, each activating the next, which eventually results in the desired effect, such as the cell dividing, or a gene being turned on. In summary, kinases work like a series of switches, controlling various processes within our cells. If something goes wrong with these "switches" – if a kinase is overly active or not active enough – it can lead to a variety of diseases, including cancer, which is why understanding kinases and their regulation is an important area of study in biology and medicine.
  • What is meant by the inline phosphotransfer with regards to a kinase?
    "In-line phosphotransfer" is a term used to describe the mechanism by which a kinase enzyme transfers a phosphate group from ATP (adenosine triphosphate) to its substrate. This is the central reaction catalyzed by kinases, enabling them to regulate a multitude of cellular processes. Before we dive into the details, let's go over a few terms: Kinase: A kinase is an enzyme that transfers a phosphate group from ATP to another molecule (the substrate). Substrate: This is the molecule that receives the phosphate group. It could be another protein, a lipid, or even another small molecule. ATP: This is a molecule that serves as the primary energy currency of the cell. One of its functions is to provide phosphate groups for phosphorylation reactions. The term "in-line" refers to the geometry of the phosphotransfer reaction. Here's what happens in simple steps: Binding: The kinase first binds both ATP and the substrate in its active site. Alignment: The phosphate to be transferred (from ATP), the oxygen that will receive the phosphate (on the substrate), and the leftover oxygen on ATP after the phosphate is transferred all align in a straight line. This is the 'in-line' part of 'in-line phosphotransfer'. This straight line is important for the chemistry to work correctly. Transfer: Once everything is aligned, the phosphate group is transferred from ATP to the substrate. The oxygen on the substrate attacks the phosphate on ATP, breaking the bond between ATP and the phosphate and forming a new bond between the substrate and the phosphate. Release: Finally, the now-phosphorylated substrate and the leftover part of ATP (now called ADP, for adenosine diphosphate) are released from the kinase. Imagine a kinase is like a factory machine, and its job is to attach a small piece (which we call a phosphate group) from an energy molecule (ATP) onto another molecule (like a protein). This attachment helps to turn the protein 'on' or 'off' and allows it to do its job in the cell. "Inline phosphor transfer" is like saying the machine uses a straight assembly line to do its job. The protein that needs the piece, the small piece itself, and the leftover of the energy molecule, all line up in a straight line. This straight line helps the machine do its job better and faster, similar to how assembly lines in factories make production more efficient. So, in summary, 'in-line phosphotransfer' is a term that describes the specific geometry and mechanism by which a kinase transfers a phosphate group from ATP to its substrate.
  • What regulates the activity of a kinase?
    In simple terms, a kinase is a type of protein that acts like a tiny machine within cells, helping to control various activities. It does this by adding a small chemical tag, called a phosphate group, to other proteins, a process known as phosphorylation. The addition of this tag can change the shape or behavior of the protein, turning its activity "on" or "off," or modulating it in some way. The activity of a kinase itself is regulated by several factors: Activation by other proteins: Many kinases need to be activated by another protein. This could be another kinase that adds a phosphate group to the kinase itself, a process called "phosphorylation." Once this happens, the kinase can do its job. Inhibition by other proteins: Just as some proteins activate kinases, others can inhibit them. These proteins work by binding to the kinase in a way that prevents it from adding phosphate groups to other proteins. Availability of ATP: ATP, or adenosine triphosphate, is the molecule that kinases use as a source of phosphate groups. If there's not enough ATP available, the kinase won't be able to do its job. Changes in the cell: Various changes within the cell can also regulate kinase activity. For example, changes in pH, temperature, or the concentration of certain molecules can all influence kinase activity. Intracellular localization: The location of a kinase within the cell can regulate its activity. Some kinases are only active when they are transported to specific regions within the cell. Expression levels: The amount of a kinase protein produced in the cell can regulate its activity. More kinase means more potential activity, while less kinase means less potential activity. In a nutshell, the activity of a kinase is regulated by a combination of other proteins, the availability of necessary resources, changes in the cell's environment, its location within the cell, and its expression levels. This intricate regulatory system ensures that the cell's functions are finely tuned and that it can respond effectively to changes in its environment.
  • What causes the mutational drug resistance of a kinase?
    Kinases are proteins in our bodies that have many important roles, including helping to control the growth of cells. Sometimes, these kinases can malfunction, leading to diseases like cancer. Some drugs are designed to target and inhibit (slow down or stop) these malfunctioning kinases, helping to control the disease. However, over time, the genes that produce these kinases can mutate, or change. These changes can cause the kinase's structure to change as well. If the structure of the kinase changes, the drug might not be able to bind to it or affect it as effectively anymore. It's kind of like trying to fit a key into a lock that's changed its shape - the key can't work if it doesn't match the lock. So, when a kinase changes due to mutation, it can become resistant to the drug that was designed to target it. This is known as mutational drug resistance. This is a significant challenge in the treatment of diseases like cancer, because it can make drugs less effective over time. Researchers are constantly studying these mutations and developing new drugs to overcome this resistance.
  • Why conformational changes occur in kinases?
    Kinases are a type of protein that play an important role in the body. They work like little machines that can turn certain processes on or off within a cell, and they do this by adding a chemical group called a phosphate to other proteins, a process known as phosphorylation. Now, these kinases don't always exist in a 'ready-to-work' state. They often need to be 'switched on' themselves. This is where conformational changes come in. A conformational change is essentially a change in shape. It's like transforming a folded up origami bird into its bird shape. In kinases, conformational changes can be triggered by the binding of molecules, such as another protein or a small molecule like ATP (the cell's energy currency). This binding usually occurs at a specific spot on the kinase known as the active site, which is like the 'working end' of the kinase. When the right molecule binds to the active site, it induces a conformational change, or shape change, in the kinase. In simple terms, this is like a key fitting into a lock, and as the key (the molecule) turns, the lock (the kinase) changes shape, becoming active and ready to perform its function. This change in shape allows the kinase to interact correctly with its target protein, adding the phosphate group to it. So, without these conformational changes, kinases would not be able to perform their job correctly. Keep in mind, it's a bit more complex than this, but this gives you a good basic understanding of why conformational changes occur in kinases.
  • What are the major types of kinase inhibitors?
    Kinase inhibitors are a type of drug that block (or inhibit) enzymes known as kinases. Kinases play many crucial roles in cells, such as helping cells grow and divide. However, when they're not working correctly, they can contribute to diseases like cancer. There are several major types of kinase inhibitors, categorized largely based on how and where they bind to the kinase enzyme: ATP-Competitive Inhibitors: These inhibitors bind to the ATP-binding site of the kinase. ATP (adenosine triphosphate) is the molecule that provides energy for the kinase's function. By occupying this site, these inhibitors prevent ATP from binding, and hence the kinase can't function. Allosteric Inhibitors: These inhibitors don't bind to the ATP-binding site, but instead bind to a different part of the kinase. This changes the shape of the kinase, which makes it harder for ATP to bind to its site and for the kinase to work correctly. Covalent Inhibitors: These inhibitors bind permanently to the kinase by forming a covalent bond. This bond is strong and irreversible, permanently blocking the kinase's activity. Non-Covalent Inhibitors: Unlike covalent inhibitors, these inhibitors form weaker, non-covalent bonds with the kinase. This means they can bind and unbind, providing a level of control that's not permanent. Each of these different types of inhibitors have their own pros and cons, and they're used to treat different diseases. In the context of cancer, for example, choosing the right type of kinase inhibitor can depend on many factors, including the specific type of cancer, the patient's overall health, and the genetic makeup of the cancer cells.
  • What is a type 1 kinase inhibitor?
    A kinase inhibitor is a type of drug that blocks or "inhibits" the action of proteins known as kinases. These kinases are very important because they help control different functions in our cells, including cell division and growth. Now, when you hear the term "Type 1 Kinase Inhibitor," it's just a way to describe a specific type of kinase inhibitor. Type 1 kinase inhibitors are special because they work by attaching themselves to the kinase at the same spot where another molecule, called ATP, would usually attach. You can think of ATP as fuel for the kinase. If ATP can't attach, then the kinase can't do its job. By blocking this ATP spot, type 1 inhibitors effectively "turn off" the kinase, and this can be useful when the kinase is causing problems, like helping cancer cells grow and divide.
  • What is a type 2 kinase inhibitor?
    A Type 2 kinase inhibitor is another type of drug that blocks or "inhibits" the action of proteins known as kinases. As we mentioned before, kinases play a crucial role in many cell processes, including cell division and growth. The difference between a Type 1 and Type 2 kinase inhibitor lies in how and where they attach to the kinase protein. A Type 1 inhibitor binds to the kinase where ATP (the energy source for the kinase) usually binds, and when the kinase is in an "active" state. A Type 2 inhibitor, however, binds to the same kinase but in a different way. It attaches itself to the kinase when the kinase is in an "inactive" state, meaning it's not currently helping the cell divide or grow. It also binds to a slightly different area, which includes the ATP-binding site but extends to an additional region called the allosteric site. The result is the same: the kinase's action gets blocked, which can be very helpful in situations where the kinase is contributing to a disease, like cancer. But the way Type 2 inhibitors achieve this is slightly different from Type 1 inhibitors.
  • What are the limitations of insilico design using AI and machine learning?
    The insilico design of kinase inhibitors, while offering significant advantages in drug discovery, also faces several limitations: Accuracy of Computational Models: The predictive accuracy of computational models is crucial. These models often rely on the quality and comprehensiveness of the data used to train them. Any inaccuracies in the structural data of kinases or their ligands can lead to incorrect predictions. Complexity of Kinase Structure and Function: Kinases have complex and dynamic structures. Their active and inactive conformations, as well as the presence of multiple binding sites, make it challenging to accurately model and predict their interactions with potential inhibitors. Lack of Experimental Validation: Insilico methods require experimental validation. Predicted kinase inhibitors must be synthesized and tested in laboratory settings to confirm their efficacy and safety, which can be time-consuming and expensive. Difficulty in Predicting Off-Target Effects: While insilico methods can identify potential kinase inhibitors, predicting off-target effects (where a drug interacts with unintended targets) is still challenging. This can lead to unforeseen side effects in later stages of drug development. Limited Understanding of Disease Context: The effectiveness of kinase inhibitors often depends on the specific disease context. Insilico methods may not fully account for the complexity of disease pathways and the role of kinases within these pathways. Computational Resource Intensity: Insilico design requires significant computational resources, especially for large-scale screenings or complex simulations. This can limit the accessibility of these methods to some researchers and institutions. Drug Resistance: Kinases can develop resistance to inhibitors, a phenomenon that is difficult to predict with insilico methods. Understanding and predicting resistance mechanisms remain a significant challenge. Integrating Pharmacokinetic and Pharmacodynamic Data: Insilico models often focus on the interaction between the inhibitor and the kinase target. Integrating pharmacokinetic (how the body affects the drug) and pharmacodynamic (how the drug affects the body) data into these models is complex but essential for accurate predictions. Data Availability and Sharing: The availability of high-quality, comprehensive datasets is crucial for effective insilico design. Restrictions on data sharing and issues with data standardization can hinder the development and validation of predictive models. Regulatory and Ethical Considerations: Insilico methods must align with regulatory requirements, which can be a moving target. Ethical considerations, especially regarding data use and patient privacy, also play a role in the development and application of these methods. At DNA SEQ, we understand the limitations of AI and machine leaning and have taken the most advantageous aspects of this technology knowing where human intersection is required to develop FDA approved drugs. The technical risk of drug design always lies with medicinal chemistry as a chief focus where we have many of the worlds foremost experts to guide this complex aspect necessary for success.
  • What does a typical pharmacophore looks like for a kinase inhibitor?
    A pharmacophore for a kinase inhibitor generally refers to an ensemble of steric and electronic features that is necessary to ensure optimal supramolecular interactions with a specific biological target, in this case, a kinase, and to trigger (or block) its biological response. These features typically include hydrogen bond acceptors and donors, aromatic rings, and hydrophobic regions. To delve into the details, a typical pharmacophore model for a kinase inhibitor usually includes: Hydrogen bond acceptors (HBA): These are primarily the backbone amide groups in the hinge region of the kinase. The hydrogen bond acceptors tend to be located in positions where they can form hydrogen bonds with the protein backbone, which is crucial for the inhibitor's ability to bind and inhibit the kinase. A common interaction of this sort involves an N-H...O=C motif. Hydrogen bond donors (HBD): These can also be part of the kinase binding motif. They are typically located in positions where they can form hydrogen bonds with the kinase hinge region. Aromatic (AR) ring groups: Aromaticity is a crucial aspect because aromatic groups can form stacking interactions with the planar residues in the active site of the kinase. They often interact with the adenine binding region of the ATP pocket and provide an anchor point for the inhibitor. Hydrophobic (Hyd) groups: Hydrophobic moieties interact with the nonpolar residues in the binding pocket, which can contribute to the overall binding affinity of the molecule. A linker or spacer group: This flexible part of the molecule connects the different functional groups and allows them to orient in a way that maximizes their interactions with the kinase. Electrostatic interaction: The phosphate binding region of the ATP pocket of kinases can accommodate charged moieties, forming strong electrostatic interactions, which are sometimes a part of the pharmacophoric features of kinase inhibitors. Inhibitors may be designed to bind to different conformations of the kinase, for example, Type I inhibitors bind to the active form, Type II to an inactive form, and Type III allosterically. Consequently, the pharmacophore models for these types may be slightly different. It's important to remember that the exact composition and configuration of a pharmacophore for a kinase inhibitor will depend on the specific kinase being targeted, as different kinases have different active sites and therefore require different features for optimal binding and inhibition.
  • How does an oncogenic kinase works and why will solving the issue of downstream signaling change the world of cancer treatment?
    Firstly, let's understand the key terms: Oncogenic Kinase: This is a type of enzyme called a 'kinase' that has become dysfunctional and promotes the development of cancer, hence the term 'oncogenic' (cancer-causing). Kinases play a crucial role in many cellular processes by transferring a phosphate group from ATP (the main energy source for cells) to specific proteins. This process, called phosphorylation, changes the function and activity of the proteins, turning various cellular processes on or off. When a kinase becomes oncogenic, it may incorrectly phosphorylate proteins, leading to the uncontrolled growth and proliferation of cells, a hallmark of cancer. Downstream Signaling: This refers to the sequence of events initiated by the activation of a protein (like a kinase) at the cell's surface. This signal is then transmitted inside the cell through various proteins, each affecting the next in a sort of relay race. Downstream signaling can lead to many different outcomes, depending on the cell type and the specific signaling pathway involved. Now, let's imagine an oncogenic kinase as a rogue switch in a highly complex electrical circuit. Normally, each switch in this circuit can turn on and off in a controlled manner, allowing electricity to flow to specific areas and power certain functions. However, if one switch malfunctions and stays permanently 'on', it can lead to a continuous flow of electricity, even when it's not needed. In the context of a cell, an oncogenic kinase is similar to this rogue switch. When it's permanently turned 'on', it continuously sends signals to proteins downstream in the pathway. These inappropriate signals can result in cells dividing and growing in an uncontrolled manner, leading to the development of a cancerous tumor. Now, why is solving the issue of downstream signaling crucial? Targeted Therapy: If we can understand and control downstream signaling, we could potentially stop or slow the progress of many types of cancer at a molecular level. Drugs could be developed to target specific points in the signaling pathway, essentially "turning off" the rogue switch or correcting the signal downstream, halting the uncontrolled growth of cells. Personalized Medicine: Each patient's cancer may involve different mutations and signaling pathways. Understanding these would allow us to create highly personalized treatments that target the specific oncogenic kinases involved in each patient's cancer. Prevention and Early Detection: Understanding these pathways could also lead to better strategies for cancer prevention and early detection. If we can identify key markers of abnormal downstream signaling, we might be able to catch cancer earlier, when it's more treatable. In essence, solving the issue of oncogenic kinase's downstream signaling can lead to significant strides in cancer treatment, potentially saving millions of lives and transforming the world of medicine.
  • Why will creating kinase inhibitors that bind to the substrate site be able to stop cancer from growing?
    Kinases are critical proteins that play integral roles in numerous cell processes, including cell growth, division, and survival, through their capacity to phosphorylate other proteins. In cancer, aberrations within kinase signaling pathways often result in uncontrolled cell proliferation and survival, contributing significantly to tumorigenesis and malignancy progression. Creating kinase inhibitors that bind to the substrate-binding site is an attractive strategy in cancer therapeutics for several reasons. The substrate-binding site of a kinase is where the target protein binds, allowing for phosphorylation to take place. By binding to this site, an inhibitor can prevent the kinase from phosphoryling its substrate, effectively blocking the signal transduction pathway and mitigating the consequential aberrant cellular behaviors. This targeted inhibition strategy, also known as competitive inhibition, can be advantageous due to its specificity. It impedes the kinase's functional capability by competing with the endogenous substrate for the same binding pocket. Therefore, its activity can be modulated by the relative concentration of the inhibitor and the natural substrate, which allows for a degree of control over the inhibition. One significant benefit of this approach is that it can minimize off-target effects commonly associated with ATP-competitive kinase inhibitors. ATP-competitive inhibitors target the highly conserved ATP-binding pocket in kinases, which can lead to unwanted inhibition of multiple kinases, causing undesirable side effects due to the lack of selectivity. Conversely, substrate sites are generally more diverse among kinases, enabling the design of highly selective inhibitors that can specifically target oncogenic kinases, thus potentially improving both the effectiveness and safety profiles of these therapeutics. However, developing substrate-competitive kinase inhibitors is not without challenges. The substrate-binding sites on kinases are often less well-defined and larger than the ATP-binding pocket, making it more difficult to design small-molecule inhibitors. Moreover, substrate-competitive inhibitors may need to overcome the often high intracellular concentrations of the natural substrate, requiring them to be extremely potent to effectively compete. Despite these hurdles, if successful, the development of substrate-competitive kinase inhibitors could offer an innovative and effective approach to disrupting cancer-associated signaling pathways, curbing malignant proliferation, and providing a valuable addition to the oncology therapeutic arsenal.
  • Explain what a well-defined substate binding site means for a substate compared to ATP binding pocket
    Let's start with some simple concepts. Substrate: A substance acted upon by an enzyme. Substrate Binding Site: It's like a "docking station" on an enzyme where a specific substrate can fit into. This is where the enzyme can perform its job, maybe breaking the substrate apart or combining it with another molecule. Each binding site is specific to its substrate, like a lock and key. Well-defined Substrate Binding Site: This is a substrate binding site that has a clear and specific structure. It's a very well-fitted "docking station", and only the right "key" (substrate) can dock properly. This ensures the enzyme is selective and only interacts with the right substrates. ATP Binding Pocket: ATP, or Adenosine triphosphate, is a molecule that carries energy within cells. An ATP binding pocket is a special type of substrate binding site specifically for ATP. The ATP binding pocket's job is to "hold" ATP so it can transfer its energy to other parts of the cell. So, it's like a specific kind of "charging dock" for the ATP "power packs". So, in short, a well-defined substrate binding site is a very specific "docking station" for its substrate. An ATP binding pocket is a specific type of these "docking stations" just for ATP, the cell's energy carrier.
  • Where the substrate site of a protein kinase is located?
    Proteins are like complicated 3D puzzles. They have lots of different parts that fold together to form a specific shape. One type of protein is called a kinase, and it's a special kind of protein that can add a small chemical group, known as a phosphate, to other proteins. This process is known as phosphorylation and can change the activity of the protein being phosphorylated. So where does this happen on the kinase? The place on the kinase where this happens is called the "active site." It's a pocket or a groove on the surface of the kinase. It's perfectly shaped so that the target protein (the substrate) can fit into this pocket, like a key fits into a lock. The "substrate site" you're referring to is simply the place in this pocket where the target protein, or substrate, binds to the kinase. This site has the perfect shape and chemical properties to allow the substrate to attach so that the kinase can do its job of adding a phosphate group to the substrate. So, in very simple terms, the substrate site of a protein kinase is the special spot where the protein that needs a phosphate group fits in, kind of like how a key fits into a specific lock.
  • Explain the potential number of DFG INTER kinases that might potentially be possible to discover.
    The number of potential DFG-Inter kinases that could be discovered depends on a few factors: Number of Kinases: In the human genome, there are approximately 518 protein kinase genes. These kinases can adopt different conformations, including the DFG-Inter conformation. Conformational States: The DFG motif (Asp-Phe-Gly) is part of the activation loop in the protein kinases, and its orientation plays a key role in the active/inactive state of the kinase. The DFG-Inter conformation is one of these states (others being DFG-In and DFG-Out). It's important to note that not all kinases may adopt all conformations, and the dynamics of these states can vary between different kinases. Diversity of Kinase Family: The human kinome (the complete set of kinases expressed in the human genome) is highly diverse. Each kinase has unique sequences, structures, and functions, leading to different regulatory mechanisms and signaling pathways. Therefore, theoretically, there could potentially be as many DFG-Inter kinases as there are kinases in the human genome (assuming they all can adopt the DFG-Inter conformation). However, it is important to note that this is an oversimplification. The actual number will depend on the structural dynamics of each kinase, their ability to adopt and maintain the DFG-Inter conformation, and the current state of our knowledge and techniques to identify and validate these conformations. Also, the development of inhibitors that selectively target the DFG-Inter conformation of these kinases is another challenge, which depends not only on the discovery of these kinases but also on drug design and development processes.
  • How would I design a pharmacophore for a DFG INTER kinase inhibitor from a research and drug discovery perspective?
    The "DFG-in" kinase inhibitors are a type of kinase inhibitors that bind to the active conformation of the kinase. The "DFG" refers to a conserved three amino acid sequence (Asp-Phe-Gly) in the activation loop of the kinase domain. Here is how one might go about designing a pharmacophore for a DFG-in kinase inhibitor. Identification of the target and structural data gathering: The first step in pharmacophore design is the identification of the target kinase. Once the target is identified, you should gather as much structural data as possible. This usually involves X-ray crystallography or NMR spectroscopy data of the kinase in complex with known inhibitors. The Protein Data Bank (PDB) is a good resource for this. It's especially valuable to find structures where the kinase is in the active, DFG-in conformation. Understanding the Binding Site: Carefully analyze the binding site of the kinase. In the DFG-in conformation, you should notice the ATP-binding pocket where most inhibitors will bind. Look for key interactions that existing inhibitors make within this pocket. The DFG motif itself should be carefully studied, particularly the position and interaction of the aspartate (D), as this residue flips in the DFG-out conformation. Initial Pharmacophore Design: Now you can begin to design the pharmacophore model. Look for common features among existing inhibitors. Hydrogen bond acceptors and donors will likely be needed to interact with the backbone in the hinge region of the kinase. A hydrophobic region or aromatic ring may be needed to fit into the adenine pocket of the ATP-binding site. A charged moiety could interact with the DFG motif itself, particularly the aspartate. Molecular Modeling and Refinement: Once you have an initial pharmacophore model, you can use molecular modeling to refine it. This might involve docking studies, molecular dynamics simulations, or other computational methods. The goal is to optimize the pharmacophore so that it has the best possible fit and interaction with the kinase. Adjust the features as necessary based on the results of these studies. Synthesis and Testing: After refinement, you can begin synthesizing compounds based on your pharmacophore model. These compounds should then be tested in vitro for their ability to inhibit the kinase. This will give you valuable information about the validity of your pharmacophore model. Iterative Optimization: Typically, the first pharmacophore model will not be perfect, and the initial compounds may not have the desired level of activity. Therefore, you should engage in an iterative process of optimization. This might involve going back to the molecular modeling stage to make adjustments based on the results of your testing. Designing a pharmacophore is a complex process that involves a deep understanding of both the biology of the target and the chemistry of potential inhibitors. It is a multi-step process that requires careful planning and execution.

DNA SEQ

7918 El Cajon Blvd, 420

La Mesa, CA 91942

© DNA SEQ 2, Inc 2024

254500_DNA SEQ_Award Logo.png

Addressing drug resistance with a novel family of DFG INTER kinase inhibitors targeting numerous cancers including; lung, breast, bladder, gallbladder, melanoma and leukemia. 

bottom of page