Issue:March/April 2025
SPECIAL FEATURE - Bioavailability & Solubility: The Promise of Novel Ingredients
There is somewhat of a consensus in life sciences that there have been significant advancements in improving bioavailability. Solubility, however, continues to elude formulators. Excipients are often lauded as a solution to tackling these challenges, but still do fall short. In a 2020 US Pharmacopeia (USP) survey of drug formulators, 84% said that the current roster of excipients present in approved drug products has imposed limitations on drug development, and as many as 28% experienced a discontinuation of drug development as a result of excipient limitations.
Novel excipients may be the answer. In September 2023, the Office of New Drugs and the Center for Drug Evaluation and Research (CDER) launched a voluntary Pilot Program for the Review of Innovation and Modernization of Excipients (PRIME). This program is intended to allow biopharmaceutical manufacturers to obtain FDA review of novel excipients. The development of novel excipients is gaining momentum as pharmaceutical companies seek improved performance and versatility in drug formulations. Novel substances support nanoparticle drug delivery for oncological medications to provide better stability and adoption of medicines.
“The invention of novel excipients bearing the amphiphilic and solubilization characteristics in the recent past has helped excipient and drug manufacturers alike to overcome the regulatory barriers for expediting the new drug candidates to market,” says Shaukat Ali, PhD, Senior Director of Scientific Affairs and Technical Marketing at Ascendia Pharmaceutical Solutions.
According to a May 2024 USP white paper: “While the FDA PRIME program represents a step in a new regulatory direction for excipients, drug developers are currently reluctant to use novel excipients as there is no independent FDA regulatory pathway outside of its drug application and approval process to review and evaluate the safety and toxicity of an excipient for introduction into a new drug, abbreviated new drug, or non-prescription drug. FDA may determine that novel excipients are not fully supported by the submitted safety data such as for the proposed level of exposure, route of administration, duration of exposure, and patient population. An entire drug application using a novel excipient could be rejected due to uncertainty surrounding acceptance of the excipient by FDA. Considering the barriers to using novel excipients that exist in the normal application process for drug products, USP supports the development of a transparent, independent approval pathway for novel excipients.”
This exclusive Drug Development & Delivery annual report explores the use of novel excipients as well as other methods and technologies for tackling bioavailability and solubility once and for all.
Abzena: Formulation Stability Study Increased Concentration
Formulating complex biologics, such as monoclonal antibodies (mAbs), fusion proteins, and bispecifics, require robust development strategies that are flexible and phase-appropriate in order to optimize them for solubility, delivery, safety, and sustained stability. Utilizing preformulation assessments to establish and characterize the physiochemical characteristics of the biologic can dictate which approaches are taken during formulation development and allow developers to rapidly identify and solve any complex problems earlier on in the process, avoiding costly downstream issues.
In a recent case study, a customer of Abzena’s had acquired a novel mAb that had been formulated by another CDMO. However, there was an issue with the selected formulation due to the fundamental properties of the mAb not being assessed during preformulation. During refrigerated storage, the formulation exhibited signs of phase separation and gelation that were resolved by warming to room temperature, without any apparent impact on quality, explains Dr. Gary Watts, Head of Formulation, Abzena.
“This was not ideal from a regulatory perspective or for patient administration,” he says. “A study was designed to evaluate the fundamental factors crucial to formulation stability, such as the optimum pH, and stability at high or low ionic strength.”
Dr. Watts says an optimal formulation was identified that showed the desired physical and chemical stability, and was clear, colorless and free of visible particles after >6 months storage at 2-8°C. Furthermore, solubility was sufficiently improved to allow the concentration to be increased to 100mg/mL from 50mg/mL, in line with the customer’s requirements.
Ardena: Exploring the Dual Role of 2-(Hydroxypropyl)-Beta-Cyclodextrin in Parenterals
Cyclodextrins (CDs) have been widely used in the pharmaceutical industry to improve aqueous solubility, bioavailability, and physicochemical stability of active pharmaceutical ingredients. Two CD derivatives are 2-(hydroxypropyl)-beta-cyclodextrin (HP-β-CD) and Sulfobutylether-Beta-Cyclodextrin (SBE-β-CD) utilized as excipients in marketed drug products. Over the past decade, CDs, especially 2-(hydroxypropyl)-beta cyclodextrin (HP-β-CD), have emerged as increasingly significant therapeutic agents mainly due to their ability to sequester and mobilize cellular lipids. Trappsol®, a proprietary intravenous formulation of HP-β-CD, is designated an orphan drug in both the US and Europe. It is currently in a Phase III clinical trial for treating Niemann-Pick Disease Type C1 and a Phase 2b study for Alzheimer’s disease. Intravenous HP-β-CD is also being studied for the treatment of diabetic kidney disease. The FDA has approved a modified gamma cyclodextrin (sugammadex®) for the reversal of neuromuscular blockade induced rocuronium and vecuronium in general anesthesia.
“These emerging applications of cyclodextrins raise an important question,” says Oluwatomide Adeoye, Formulation Development Scientist, at Ardena. “If these ‘excipients’ exhibit therapeutic activity, is their continued classification as pharmaceutical excipient justifiable, given that they are traditionally expected to be inert? Our experience as a CDMO developing New Chemical Entities (NCEs) for various pharmaceutical and biopharmaceutical companies indicates a growing industry concern regarding the use of cyclodextrins as solubilizers in new parenteral drug products.”
Timothy Pas, Director, Formulation Development & Production at Ardena, adds that these concerns primarily stem from the risk of ototoxicity associated with high concentrations of HP-β-CD observed in clinical studies for Niemann-Pick Disease Type C1. “While the causative mechanism of HP-β-CD induced ototoxicity has not been perfectly elucidated, the consensus is that it is related to altered lipid trafficking in the cochlear structures,” says Mr. Adeoye.
The long history of use in parenteral formulations suggests that the risk of HP-β-CD induced ototoxicity is minimal at typical concentrations and parenteral use cases, Mr. Pas says. However, formulation scientists must carefully assess the clinical requirements of a drug before selecting HP-β-CD as solubilizer. “For instance, it is reasonable to avoid its use in drug formulations intended for prolonged administration, where a regular maintenance dose is required, or those that may lead to high cumulative concentrations of HP-β-CD,” says Mr. Adeoye. “Alternatively, SBE-β-CD should be the preferred choice for drug formulations when it can form non-inclusion complexes with the same drug, as its reduced interaction with cell membranes minimizes the risk of cholesterol sequestration.”
Ascendia Pharmaceutical Solutions: Solubilization Technologies Tackle Challenging Molecules
As more molecules coming out of discovery possess poor solubility, industry is relying on more innovative approaches to enhance the permeability and bioavailability of those molecules. Excipients are crucial to help design better and smarter formulations to achieve the desired solubility to overcome the bioavailability challenges. Compounded with stability challenges, the industry is looking for the ingredients, solubilizers, and polymers to maximize the drug-polymer intermolecular interactions in the formulation matrices.
“Ascendia’s enabling solubilization technologies, especially EmulSol®, address the challenging molecules for enhancing the solubility and oral bioavailability of drugs formulated in microemulsions and nanoemulsions comprised of lipid-based solubilizers and surfactants, and oils as well,” says Shaukat Ali, PhD, Senior Director of Scientific Affairs and Technical Marketing at Ascendia Pharmaceutical Solutions.
Lipophilic drug molecules (BCS II and IV) require the most advanced technologies to improve their solubility and bring them to the clinic. For instance, the molecules having higher melting and log P due to their inherent high lattice energy and lipophilicity with log P (4-8), require solid amorphous dispersion and/or microemulsion technologies.
“Ascendia’s enabling solubilization technologies AmorSol® for oral solids and EmulSol and NanoSol® for oral liquids can help engineer better and smarter formulations for tablets and liquid capsules using generally regarded safe (GRAS) excipients approved by FDA,” he says. “These technologies are aimed at improving solubility of lipophilic and hydrophobic molecules bearing higher melting and logP. Designed with a carefully selected class of excipients and lipids/surfactants/solubilizer, and coupled with our expertise in the enabling technologies, we can expedite the formulation and development of molecules and bring them to clinic faster.”
Excipient/surfactant composition and particle size also have a great impact on formulation composition and the desired outcomes. For instance, cyclosporine A available as Sandimmune® (comprised of corn oil, linoleoyl macrogolglycerides) is prone to much greater food effect than Neoral® formulation (comprised of corn oil, polyoxyl 40 hydrogenated castor oil) with much smaller particle size. Dr. Ali says that Ascendia’s Emulsol technology can be used for drugs like cyclosporine and peptides to achieve faster absorption and permeation, thus higher oral bioavailability.
BioDuro: Exploring Cutting-Edge Tech for More Reliable Formulations
The development of enhanced drug formulations requires an in-depth, data-driven understanding of Active Pharmaceutical Ingredients’ (APIs) physicochemical properties and their behavior in vivo. At BioDuro, formulation challenges such as poor solubility, stability, and bioavailability are addressed by systematically analyzing key characteristics of the API and the target patient population, explains Dr. Yuan Yang, Senior Scientist III, Formulation Development, BioDuro. A scientific approach that balances the inherent risk of formulation complexity with the practical needs of the drug product is critical for success.
For APIs with poor solubility, several formulation strategies are employed, each with specific advantages and challenges depending on the solubility profile and therapeutic window. Nanoparticles, nanocrystals, lipid-based formulations, cyclodextrin incorporation, and Amorphous Solid Dispersions (ASD) have proven to be essential tools for solubility enhancement. “BioDuro specializes in employing these strategies, selecting the most suitable formulation based on factors such as API stability, solubility, partition coefficient, and the required dose strength,” he says.
By transforming the API into an amorphous state and dispersing it in a polymer matrix, ASDs improve dissolution rates while maintaining stability. This strategy is particularly beneficial for drugs with poor solubility, as it enhances dissolution rates and improves the drug’s absorption, leading to more consistent and predictable bioavailability. By leveraging precise modeling and predictive tools, formulation scientists can mitigate risks while maximizing therapeutic performance.
The core of formulation enhancement often lies in the ability to manipulate the physical form of the API. Spray drying and hot melt extrusion (HME) are widely used, proven techniques that provide effective solutions for API solubility challenges. Conventional spray drying has been the standard for producing amorphous dispersions and enhancing the bioavailability of poorly soluble drugs. Dr. Yang says: “With a success rate of over 90% in scale-up from lab to commercial manufacturing, spray drying is a reliable method for formulating high-quality solid dispersions.” HME, similarly, is recognized for its capacity to produce stable, uniform formulations and is integral to the development of immediate-release and controlled-release dosage forms, with more than 50% of commercial products utilizing this technique. However, the increasing complexity of drug candidates has led to the exploration of newer technologies to address evolving challenges. “Electrospray, a cutting-edge technique, has gained significant attention in producing ASDs,” he says. “Electrospray offers several advantages, such as highly repeatable and reproducible processes and the ability to produce fine, uniform particles with a narrow size distribution. Studies show that electrospray-generated formulations exhibit improved stability and dissolution rates compared to conventional methods, making them particularly beneficial for high-potency, low-dose drugs.”
In addition to electrospray, other emerging technologies are being explored to enhance solubility and bioavailability. Techniques, such as formulating with solubility-enhancing excipient, amorphous dispersion granulation and coating fluid bed, co-precipitation, and electrospinning are being evaluated for their potential to improve the solubility of challenging APIs. These approaches, alongside spray drying, HME, and electrospray demonstrate the pharmaceutical industry’s ongoing commitment to innovation. “BioDuro continues to explore and implement these cutting-edge techniques to develop more efficient and reliable drug formulations,” says Dr. Yang.
Bend Bioscience: Spray Drying Is Best in Early-Phase Design
As a CDMO specializing in bioavailability, particle engineering, and solubility enhancement via spray drying, Bend Bioscience has tackled complex formulations for decades. “We find that the spray drying process is best for early phase design work in terms of material sparing, speed, and efficiency,” says David Vodak, PhD, Chief Scientific Officer, Bend Bioscience, Oregon. “We are, however, still observing that amorphous solid dispersion (ASD) technology is the most applicable to a broad range of solubilization problem statements. Certain combinations of ASD technology with other functional architectures, like controlled release, seem to be trending with respect to dialing in specific release and extent profiles for challenging molecules and target product profiles.”
Understanding the relative release rates of functional excipients and active molecule are important features to achieve optimal performance, and designing a particle architecture through spray drying can be enabling, he says. “All molecules are unique and have specific challenges to deliver a successful commercial dosage form. Our science-based approach to design – incorporating a deep understanding of material science, rigorous experimentation and analysis, and emphasis on process optimization and scalability – translates to efficient and effective progressable solutions in drug development.”
Lipophilic (LogP>2) compounds remain prevalent in Bend’s portfolio. Many compounds in the kinase inhibitor class tend to be more polar with high melting points (i.e., ‘brick dust’). The company is seeing high logP compounds in trends toward PROTACs and other TPD programs. Although these compounds are lipophilic, Dr. Vodak says they are often ionic and higher molecular weight moving them into the BCS IV regime.
“At Bend Bioscience, we are still working to discover a ‘magic bullet’ in terms of the technology to delivery these types of molecules,” he says. “In the meantime, we are able to efficiently apply our experience and acumen to design formulations that solve the challenge of delivery in a commercializable manner, whether a spray dried amorphous dispersion or lipid-based formulation is optimal.”
Artificial Intelligence (AI) is also helping Bend formulate those challenging molecules. AI is helping Bend organize data sets and look for patterns in compound properties and successful formulations. “We have evaluated ‘black box’ modeling and deep learning algorithm techniques, but have not yet found a satisfactory data set with appropriate truth metrics to ensure anything predictable from the data,” says Dr. Vodak. “However, it is early days for AI, and we fully expect to stay on the cutting edge in this area and use the data that we have from a diverse set of pipelines to continue to challenge what AI can do to improve our efficiency in formulation and process design.”
Catalent: PBPK Modeling Identifies Root Causes
Developability assessment with physiologically-based pharmacokinetic (PBPK) modeling has transformed Catalent’s early development service. “The use of PBPK modelling replaces simplistic tools like BCS/DCS classification, that oversimplify the challenges in bringing a molecule to the clinic for simple small molecules as well as those in the Beyond Rule-of-5 space,” says Stephen Tindal, Director, Scientific Advisory, Oral Small Molecules, Catalent.
As an example, a small biotech faced bioavailability and dose escalation failures with a Beyond Rule-of-5 molecule, leading them to consider shelving the project. Instead of trial-and-error formulation, Catalent applied its Developability Assessment, integrating PBPK modeling and early development screening to identify the root cause.
“Oral delivery proved unviable, so we recommended a sublingual formulation that significantly improved systemic exposure,” he explains. “We also optimized their preclinical study design by identifying species-specific absorption issues and developed a modified-release strategy to manage dose escalation. This structured approach saved the molecule from termination, accelerating its path back to the clinic and reducing development costs.”
He adds that Catalent’s expertise in technology selection, route evaluation, and formulation design empowers biotech teams to make data-driven decisions, salvaging high-risk molecules and optimizing early development.
Croda Pharma: Enhancing Drug Formulation with Machine Learning
Drug formulation that exhibits enhanced solubility, stability, and bioavailability is a challenging craftsmanship. In recent years, many have tried resorting to Artificial Intelligence (AI) and Machine Learning (ML) to streamline this process. “Unfortunately, developing digital solutions for drug formulation is difficult by itself, with challenges such as low amounts of (relevant) data points, the need to model complex systems, and frequently changing objectives that require practitioners to shoot at moving targets,” says Claas Strecker, Lead Technical Data Scientist, Croda Pharma.
Croda aims to empower the drug formulation process with smart digital approaches that can support drug developers in navigating complex systems, focusing on three key pillars:
1) Automatization: Automating tedious lab experiments is of core importance. “It is not only improving the productivity of our scientists and allowing us to respond to our customers faster, but also opening access to vaster amounts of data that can fuel more complex predictive models downstream,” explains Mr. Strecker.
2) Predictive Modelling: Croda has built a model that predicts if combinations of excipients will form stable self-(micro)emulsifying drug delivery systems (S(M)EEDS). “These systems have emerged as a powerful tool in pharmaceutical formulation and enhance the solubility and bioavailability of poorly water-soluble drugs by spontaneously forming fine oil-in-water emulsions in the gastrointestinal tract under mild agitation, thereby improving drug dissolution and absorption,” says Veronica Blanco, Lead Applications Scientist, Croda Pharma.
Despite their potential, optimizing S(M)EDDS formulations is a complex, iterative, and time-intensive process. Key variables, such as the selection and ratio of excipients, drug characteristics, and system stability, require extensive experimentation. Ms. Blanco says: “Thus, we challenged ourselves to accelerate this experimentation by using ML, but quickly faced a major challenge: How could we inform our model to learn the underlying physics of such complex systems?”
Excipients such as polysorbate 80 are not one well-defined compound, but constitute structurally diverse mixtures. Moreover, Croda’s series of super-refined (SR) excipients only deviate from their regular counterparts at the trace component level, but exhibit significantly improved formulations properties, says Mr. Claas. “During our work, we found that NMR (nuclear magnetic resonance) spectra can serve as a good feature source to inform our predictive model.”
3) Optimization: Drug formulation predictive modelling frequently falls short either simply due to lack of relevant data to tackle the problem at hand or because, for complex systems, no informative descriptors are found that could inform a predictive model sufficiently enough, explains Mr. Claas. In such scenarios, optimization techniques such as Bayesian optimization (a method for finding the best solution to a problem without in-depth understanding of how it works internally) can be used. Different options are evaluated, results are presented, and well-calibrated probabilities of expected improvements are used to make smarter choices. “This can help us to iterate fast and achieve our objectives without wasting vast amounts of resources and time,” says Mr. Claas.
Ms. Blanco adds: “As formulations continue to become more complex, automation improves efficiency and data collection, while predictive models help design stable drug delivery systems.”
Gattefossé: Lipid Excipients Enhance Drug Absorption
Lipophilic drug compounds are a major component of the pre-clinical pipeline, and formulators are increasingly seeking approaches to enhance absorption and drug permeability. A PharmaCircle analysis performed by Gattefossé showed that approximately 70% of NCEs in the preclinical pipeline today are poorly soluble. However, only 55% of FDA-approved NCEs from 2019-2023 are considered poorly soluble compounds. “This 15% gap shows that traditional solubility enhancement techniques are not sufficient to bring all these challenging new molecules to market,” says Nick DiFranco, Senior Marketing Manager – Pharmaceuticals, Gattefossé USA. “There is significant untapped potential for lipid-based drug delivery systems, which not only address poor solubility but also enhance permeability and absorption of challenging compounds.”
An area of growing interest is utilizing combinations of excipients to access new “synergies” and overcome common formulation challenges. “For example, an amorphous solid dispersion (ASD) may solubilize a high dose of API, but these formulations often face in vivo performance and stability challenges,” says Mr. DiFranco. “In contrast, lipid-based formulations (LBFs) improve drug permeation and absorption through mechanisms such as tight junction modulation and lymphatic uptake. LBFs may also reduce dosing variability associated with the food effect.”
Lipid excipients significantly improve drug delivery by enhancing the permeation and absorption of drugs. Their similarity to dietary lipids allows them to be processed through the digestive system, increasing the solubility and supersaturation of lipophilic drugs, especially for BCS Class II and IV compounds. The lipid digestion process, involving gastric lipase, bile salts, and pancreatic enzymes, also creates colloidal carriers and mixed micelles that prevent drug precipitation. These colloidal structures maintain drug solubility and can penetrate the mucus layer, releasing the drug and creating a supersaturated environment for passive diffusion.
“By combining LBFs and polymeric formulations, scientists can achieve the high drug loading of an ASD with the benefits of lipids, such as improved in vivo permeability and prevention of recrystallization, resulting in boosted bioavailability,” he says. “Utilizing IID-listed polymers and lipids in these ternary systems ensures innovation without additional regulatory risk, and these systems can often be manufactured with existing equipment, ensuring a clear path to scale-up and commercialization.”
Excipients with high medium-chain fatty acid ester content, such as Gattefossé’s Labrasol® ALF, Capryol® 90, and Labrafac™ MC60, are especially good at modulating tight junctions, enhancing paracellular permeation for Class III and IV compounds and helping drugs bypass P-glycoprotein (PgP) inhibition, says Mr. DiFranco.
As an example, alendronate sodium, a BCS Class III drug, has very low bioavailability due to its polar, hydrophilic nature and susceptibility to pH and enzymatic degradation. In an in vivo rat study using a closed-loop small intestinal administration, vehicles with a high medium-chain fatty acid ester content, namely Capryol 90, Labrasol ALF, and Capryol PGMC, were the most effective permeation enhancers (Ukai et al. 2020). Capryol 90 generated 9.8-fold increase in AUC, which was attributed to an increase in both transcellular and paracellular uptake facilitated by intestinal membrane fluidization and safe, reversible modulation of tight junction proteins.
Additionally, a human clinical study on enlicitide chloride, a macrocyclic peptide developed by Merck & Co., demonstrated the ability of Labrasol ALF to improve solubility and modulate tight junctions, generating a 2- to 3-fold increase in plasma concentration (Johns et al. 2023).
“These studies show how lipid excipients with high medium-chain fatty acid ester content, such as Capryol 90 and Labrasol ALF, can greatly enhance drug absorption in vivo, especially for BCS Class III and IV compounds,” says Mr. DiFranco.
Hovione: Early-Stage ASDs by Spray Drying Result in Viable Oral Dosage Form
Amorphous solids dispersions (ASDs) remain the most used enabling platform for solubility enhancement. There are a variety of molecule structures in the pipeline that require tailored bioenhancement, including the common greaseballs as well as chameleonic and brick-dust APIs. “ASDs by spray drying are a versatile platform to formulate across the board,” says Inês Ramos, R&D Manager (Formulation Development, Oral Drug Product) at Hovione.
For BCS II and IV compounds, bioavailability enhancement is driven by absorption enhancement needs, target product profile, and drug molecular features. “To maintain a strong focus on manufacturability since early-stage, the delivery of a viable ASD oral dosage form follows an integrated approach involving ASD formulation screening, particle engineering, drug product formulation and process development using data-driven tools to expedite development,” says Dr. Ramos.
Hovione’s approach relies on a streamlined workflow supported by computational tools that starts with a “technology fitting” to assess the suitability of using ASDs by spray drying. ASD development includes a comprehensive high-throughput formulation screening (ASD-HIPROS™ proprietary platform that includes common polymers and alternative excipients such as the Dispersome® technology), designed to fast-track first-in-human (FiH) formulations that are scalable and provide adequate performance. The ASD-HIPROS platform requires a few grams of API and less than eight weeks to narrow down thousands of possible formulations to the most viable candidates. The drug product intermediate is then formulated into a tablet, capsule, or pellets/granules for oral delivery. “This methodology was designed to expedite the delivery of an enabling formulation and an industrially viable manufacturing process,” says Dr. Ramos. “The goal is to maintain performance and ensure patience compliance.”
LATITUDE Pharmaceuticals: Formulating the Most Insoluble Compounds
LATITUDE Pharmaceuticals takes a pragmatic approach to identify the most effective solubilization technologies for each client’s individual desired drug profile. For more potent compounds with an estimated human dose under 200mg, the company utilizes amorphous solid dispersions and self-emulsifying formulations, which are free from synthetic lipids, detergents, and organic solvents – ensuring improved safety profiles and compatibility with capsule shells, says Andrew X. Chen, President and Founder of LATITUDE Pharmaceuticals. For drug substances with an estimated human oral dose exceeding 200mg, the company typically develops nanoparticle suspensions or tablet formulations containing API nanoparticles, which significantly enhance the dissolution of insoluble compounds.
“For injectable formulations, we have successfully formulated even the most insoluble compounds using one of our three preferred solubilization technologies: nanosuspensions, nanoemulsions, and LATITUDE’s own ClearSol™, all of which are free from toxic or allergenic ingredients, and only contain components approved by the FDA for injection,” he says. “Any of these formulations are readily scalable and can be manufactured under GLP or GMP conditions for human use.”
Over the past 12-18 months, Dr. Chen says roughly 50% of the projects that LATITUDE developed were for compounds that were lipophilic, insoluble, and required increased bioavailability. LATITUDE often relies on a combination of a bile acid with triglycerides and lecithin (as in its patented ClearSol solubilization platform) for significantly enhanced solubility and safety. “ClearSol is a highly effective, broad-spectrum solubilizer with a systemic and local safety profile that is comparable to, and often better than, cyclodextrins,” he says.
Nanoform: Reduce Pill Regimens with Nanoforming
While numerous biologics have been approved in the last decade, small molecules still dominate development pipelines. As the complexity and lipophilicity of new APIs steadily increase, the challenges associated with poor solubility, leading to incomplete absorption and suboptimal pharmacokinetics, persist.
Established technologies such as ASD, nanomilling, cyclodextrins, and lipid formulations have been extensively reviewed. Of the emerging technologies, Tamas Solymosi, PhD, Lead Scientist and Technical Sales Manager, Nanoform, says it is worth mentioning deep eutectic solvents capable of solubilizing poorly soluble compounds, drug-drug co-amorphous systems improving both the solubility and stability of APIs even compared to individual amorphous states, and nanoforming, which is a green, ‘bottom-up’ particle engineering process to produce nanocrystalline or nanoamorphous particles.
Nanoforming technology can decrease the particle size of APIs to the 10nm range. The process works by the controlled precipitation of APIs from supercritical CO2 (scCO2) solution. “As nanoforming is relatively new, it is necessary to establish whether a compound is suitable for the nanoforming process,” says Dr. Solymosi. “Nanoform uses its sparse data AI tool, STARMAP®, to predict the solubility of an API in scCO2. This enables formulators to select the best suited APIs before committing to experimental work.”
Nanoforming can also be used to enable single pill-per-day regimens for numerous poorly soluble drugs. Consider that early formulations sometimes sacrifice patient-centric perspectives with large pill burdens and strict food restrictions. He says: “Reformulations and optimized compositions can help improve patient centricity and adherence to therapy.”
Dr. Solymosi adds that initial lipid-based capsules are typically phased out by ASDs. “We have seen that for enzalutamide, decreasing pill size and pill burden.”
Nanoform successfully nanoformed the prostate cancer drug enzalutamide, already marketed as an ASD. The marketed treatment has a regimen of 4x40mg pills per day. By formulating the treatment in a nanocrystalline form, a single 160mg pill regimen was developed that matched the exposure of the supersaturating ASD formulation. Production was scaled up to several tons/year and the nanoformed tablet showed promising results in clinical evaluation and is set to enter the market in 2027.
Pharmaceutics International Inc.: Physical Property Evaluations Predict Solubility
In recent years, there has been a lot of focus on biologics and personalized medicine that are mostly delivered via parenteral routes, but oral medication is still the most convenient route of administration accounting for more than 80% of approved drugs. Use of combinatorial chemistry and high throughput screening in pharmaceutical discovery for the next drug candidate has kept funneling on molecules with poor aqueous solubility. Ninety percent of the new chemical entities and 75% of the compounds under development have inadequate water solubility. Biopharmaceutical Classification System Class II and IV compounds fall into this category. Bioavailability of an active moiety, among other factors, is predominantly affected by its solubility and permeability. Adequate aqueous solubility at absorption site is necessary for sufficient permeation and pharmacological action translating to desired clinical outcome.
To achieve optimal bioavailability, scientists have used various physical and chemical approaches to modify the rate of dissolution and solubility. Prodrug, salt formation, and cocrystals are the main chemical approaches that are used in early development to enhance solubility. Physical approaches to improve solubility have been mainly focused on solid dispersion, lipid-based drug delivery systems using oils, lipids, surfactants and/or cosolvents, particle size reduction and Cyclodextrin inclusion complex. Primary technologies that were used to improve solubility in FDA-approved products since 2000 are hot melt extrusion and spray drying to form solid dispersion; self-emulsifying -emulsion, -microemulsion, and -nanoemulsion systems (SEDDS, SMEDDS, SNEDDS) for lipid-based delivery; micronization and nanoparticles using dry or wet milling processes; and Cyclodextrin inclusion complexes. Among these, 30% of products were based on a solid dispersion platform.
At Pharmaceutics International Inc., a Jabil Company, initial evaluation of physical properties of insoluble drug molecules is done utilizing Hansen Solubility Parameters in Practice (HSPiP) software to predict the solubility in various solvent or solvent combinations, explains Sundeep Sethia, Vice President of R&D at Pharmaceutics International.
“Here, the API and solvent are characterized by just three parameters δD for Dispersion (van der Waals), δP for Polarity (related to dipole moment) and δH for hydrogen bonding and represented as points in three dimensional HSP space,” he says. “API and solvents sharing similar HSP space shows favorable solubility. Solubility is subsequently verified by experimental data. Based on the output, a typical formulation approach may be development of a SEDDS or SMEDDS using lipidic excipients and delivered as an oral liquid or soft gelatin capsule dosage form. Softgel capsules mask the oily taste and provide ease of unit dosing.”
For compounds with poor solvent solubility, wet milling to nano size to increase the diffusional surface area is one option. Here, the API is milled along with stabilizers (surfactants/co-surfactants) using mills, such as agitator bead mill (Dyno®Mill), high pressure homogenizer, etc. Mr. Sethia says that increased surface area improves in vivo dissolution, thereby improving bioavailability.
Quotient Sciences: SDD Suspension Enhances Absorption
Dr. Vanessa Zann, Vice President, Scientific Consulting, Quotient Sciences, says she has seen positive results with spray dried dispersion (SDD) formulations to help enhance fraction absorbed, which in turn increases exposure. Quotient Sciences uses biorelevant dissolution to rank solubility-enhanced technology platforms and then selects the most promising for clinical assessment.
“Note there is often a disconnect between preclinical in vivo data and clinical outcomes,” she says. “We recommend assessing the SDD platform initially as a suspension formulation, which provides gold standard data for what is likely to be achieved with a good performing SDD tablet.”
For example, Tranquis approached Quotient Sciences in the preclinical stage to help develop clinical formulations for first in human (FIH). The compound in question was a BCS II compound (poor solubility and high solubility) and had shown solubility limited exposure preclinically, hence an enhanced formulation was anticipated to be required to achieve efficacious clinical exposures, explains Ms. Zann. Quotient Sciences used an integrated approach for drug product development, manufacturing, and clinical testing (this is the Translational Pharmaceutics® platform) to rapidly identify and overcome solubility and bioavailability challenges for this compound.
Three suspension formulations were developed for assessment in the FIH: a methylcellulose crystalline (MC) suspension; a spray-dried dispersion (SDD) suspension; and a hot melt extrusion (HME) suspension. The regulatory package was filed with 90 days stability for the SDD and HME GMP intermediates and seven days for the powder in bottle (PiB). The suspension had four hours in use stability and a rinsing trial performed on each to allow total dosing volume to be 240mL. She explains that the SDD and HME both showed superior dissolution to the crystalline API in a biorelevant dissolution assessment, demonstrating a spring and parachute effect with reduced precipitation in the intestinal phase. A number of SDD suspensions were also assessed in the rat prior to the FIH but didn’t show any improvement over the MC suspension.
The FIH study started dosing SAD cohort 1 (60mg) and 2 (180mg) with a methylcellulose crystalline suspension, which gave three-fold higher exposure than anticipated from the preclinical species. At SAD cohort 3 (540mg), the subjects were initially dosed with the MC suspension, but then returned to the clinic for dosing with the SDD and HME suspensions. The dose of the SDD and HME suspension was reduced to 180mg to ensure that the exposure caps were not exceeded due to the already higher exposure in the clinic than originally predicted. The SDD had the highest exposure showing a four-fold increase compared to the MC suspension. This formulation was selected for dosing the remainder SAD and MAD cohorts.
“The Translational Pharmaceutics platform allowed the study not only to deliver safety, tolerability, and pharmacokinetic data, but also formulation selection assessment ahead of Phase II patient trials,” says Ms. Zann.
Serán Bioscience: A Scientific Approach to Understanding a Molecule’s Barriers
The landscape for oral delivery of novel new chemical entities (NCEs) is rapidly evolving as we leave the “rule of five” era, says Rod Ketner, PhD, Vice President of Business Operations at Serán Bioscience. Increasingly large and complex “small” molecules with poor solubility and permeability present unique challenges to drug delivery. Successful formulation strategies must meet the target product profile with a robust and scalable manufacturing process, often on accelerated development and approval timelines.
Serán’s team takes a hypothesis- driven scientific approach to understanding the physical, chemical, and biological barriers to absorption. “Avoiding a shotgun approach to formulation design, we characterize an NCE and its performance in biorelevant media and dissolution conditions,” he says. “An understanding of a molecule’s performance in these media can set the foundation for formulation approaches and dosage form architecture.”
Conventional formulation strategies – such as leveraging the low-pH environment of the stomach to solubilize weak bases – increasingly prove insufficient and more advanced techniques are necessary, he says. Converting crystalline APIs into amorphous solid dispersions (ASDs) can substantially increase solubility, but require excipients that sustain supersaturation through GI transit. “ASDs, manufactured through methods like spray drying, melt extrusion, or precipitation, yield a higher free energy state and, consequently, greater solubility in the small intestine,” says Dr. Ketner. “Amorphous forms have been demonstrated to lead to a tenfold or greater enhancement in solubility.”
Spray dried dispersions (SDDs) stand out as a preferred method for enhancing solubility in the gastrointestinal (GI) tract. These stable, amorphous formulations combine an API and a polymer in an organic solvent, followed by spray drying to create particles ranging from 5-50um. The resulting particles exhibit physical stability and dissolution properties that significantly boost bioavailability compared to crystalline API alone. “The scalability and well-established nature of spray drying technology are notable, with over 50 approved SDDs on the market currently,” he says. “However, developing and scaling SDDs effectively requires specialized experience, not just an off-the-shelf dryer.”
Beyond solubility, permeability-limited absorption is an increasing obstacle, especially for complex molecules like protein degraders and peptides. Their larger size (800-1500 MW) reduces their passive permeability across the epithelium in the small intestine, and complex self-assembly can lower the driving force for permeation. Multiple approaches have been screened to address these issues, including lipid systems, and additional permeability enhancers. Permeation enhancers that modify the lipid bilayer or tight junctions between epithelial cells remain controversial and require further research.
“Given these challenges, outside-the-box approaches to addressing permeability challenges are essential for achieving acceptable pharmacokinetics,” says Dr. Ketner. Nanoparticles, for instance, can maximize drug concentration at the surface of the epithelium. For peptides, protecting them from enzymatic degradation in the GI tract and designing dosage forms that transit, protect, and dissolve at the right location is critical. Patient-friendly injectable options, such as subcutaneous and intramuscular formulations, also merit consideration for drugs facing significant permeability issues.
Serán applies materials science principles to develop formulations, including enabled approaches like ASDs, along with scalable processes designed for sustained progress. He says: “This allows Serán’s team to offer flexible formulations and processes that support evolving clinical study needs and ensures R&D resources are effectively focused on the assets most likely to be successful.”
Spokes Sciences: Highlighting the Utility of SNAC with Lipophilic Compounds
In both the development of drug products for new chemical entities (NCEs) and within product life cycle management of existing drug products (such as developing oral dosage forms out of known actives currently administered by injection), major hurdles to the use of new pharmaceutical excipients are the time, cost, and uncertainty associated with obtaining FDA approval. Despite lipophilic drug compounds remaining a driving force behind the development of bioavailability enhancement technologies, these regulatory approval challenges can hamper or delay the adoption of novel functional excipients for that purpose, explains Kimberly Zubris, PhD, Chief Science Officer, Spokes Sciences.
Originally developed to improve the oral bioavailability of water-soluble active ingredients, salcaprozate sodium or SNAC, is an absorption-enhancing excipient already approved by the FDA and used in a blockbuster diabetes drug with the active ingredient semaglutide (Rybelsus®). SNAC’s generally recognized as safe (GRAS) status means it could immediately be used in over-the-counter supplements, says Dr. Zubris.
Spoke Sciences creates innovative proprietary technologies through the delivery of highly lipophilic/poorly water-soluble active pharmaceutical and plant-derived functional ingredients. Previously, Spoke and its predecessor discovered that SNAC could be combined with some lipophilic functional ingredients to make new proprietary oral dosage forms with significantly improved bioavailability. Striking among these data are a human clinical study showing that SNAC increases cannabidiol (CBD) oral bioavailability by 6-fold compared to standard solid-filled capsules without SNAC, and another clinical study showing that SNAC increases oral tetrahydrocannabinol (THC) exposure by 2.3-fold compared to THC in ethanol.
Highlighting the utility of SNAC as a blockbuster absorption enhancing ingredient, Dr. Zubris says that Spoke has recently undertaken studies to show that a broad range of hydrophobic actives can be combined with SNAC to significantly improve both their aqueous solubility and their rate of dissolution. This SNAC-driven solubility enhancing effect applies to molecules that span the range of the lipophilic spectrum, including: melatonin (Log P ≅ 1.6), resveratrol (Log P ≅ 3.09), pterostilbene (Log P ≅ 3.54), ibuprofen (Log P ≅ 3.8), curcumin (Log P ≅ 4.28) and coenzyme Q10 (Log P ≅ 10). With a stoichiometric optimum ratio of SNAC:active, solubility was seen to increase by up to 350 times in the presence of SNAC relative to that of water alone.
“SNAC enhances the oral bioavailability of lipophilic drugs, not just hydrophilic drug substances as previously assumed,” she says.
Ongoing studies at Spoke continue to develop data validating SNAC’s mechanism of action with these lipophilic active ingredients. But given these results, Dr. Zubris says that SNAC is well poised to become a key tool for the use in formulating poorly absorbed drugs.
Thermo Fisher Scientific: Molecule-Centric Approach to Amorphous Drug Development
When asked how Thermo Fisher Scientific solved a bioavailability issue for a client, Sanjay Konagurthu, PhD, Senior Director, Science and Innovation, Pharma Services, at Thermo Fisher Scientific, shares the design of a solubilized amorphous spray dried dispersion (SDD) that enhanced oral bioavailability of a poorly water-soluble drug. To support this partner, Thermo Fisher used a molecule-centric approach to amorphous drug development that, instead of empirical or ‘shotgun’ approaches to accelerate formulation screening, is routinely performed leveraging Thermo Fisher’s Quadrant 2® approach. Quadrant 2 is an artificial intelligence (AI) and machine learning (ML)-based predictive modeling platform, designed for solubility and bioavailability enhancement.
“Lead formulations were identified with a combination of in silico modeling and in vitro solubility-supersaturation measurements to predict drug-excipient interactions,” he explains. “The combination of computational and experimental tools provides a cost- and time-efficient strategy to select drug-excipient combinations, prior to embarking on more time-consuming formulation and process development.”
Thermo Fisher performed molecular modelling to characterize interactions of pure drug, excipients, and drug-excipient systems via molecular dynamic (MD) and quantum mechanical (QM) simulations using a suite of programs assembled by the team. The goal of this work was to examine the drug-drug and drug-excipient molecular level interactions between a BCS Class II molecule and compendial/GRAS polymers, in order to provide a rational basis for the selection of appropriate polymers for inclusion in a solubilized drug product intermediate.
“This rationale is based on molecular descriptors and specific drug-polymer interaction energies. In silico modelling results were corroborated by an in vitro solvent spike assay,” Dr. Konagurthu says.
Lead drug loading and polymer systems were characterized by in vitro tests and confirmed by in vivo animal studies. In agreement with the in silico predictions, SDDs demonstrated substantial improvement of bioavailability in both in vitro and animal pharmacokinetic studies compared to the crystalline drug. Solvent spike assay analysis demonstrated that the addition of polymer excipients improved the sustainment of supersaturation in FaSSIF (Fasted State Simulated Intestinal Fluid). Non-sink dissolution results agreed with predictive in silico modelling and solvent spike analysis that HPMCP HP-55, Eudragit L-100 and CAP provided the greatest synergistic effects and provided improved sustainment of supersaturation.
He says: “The in vivo data demonstrated that the SDDs resulted in significant enhancement of oral bioavailability. SDDs with a ranging polymer composition and drug loading provided up to an approximately three-fold increase in maximum drug plasma concentration and an approximately seven-fold increase in total AUC (Area Under the Curve). In silico modeling accurately predicted that HPMCP HP-55, CAP and Eudragit L-100 based SDDs would provide miscible drug-polymer systems with significantly enhanced performance.”
In summary, Dr. Konagurthu says that a combination of experimental and computational modeling approaches allows for predictive drug-polymer miscibility and supersaturation sustainment. “Utilization of Quadrant 2 modeling allows for rapid screening with zero Active Pharmaceutical Ingredient (API) usage and a significant reduction in benchtop manufacturing/analysis,” he says. “This led to the successful development of an enabling formulation for a BCS Class II drug that demonstrated superior oral bioavailability by leveraging our Quadrant 2 platform.”
Total Page Views: 1009