Feeds:
Posts
Comments

Posts Tagged ‘advances in drug development’

Live Notes, Real Time Conference Coverage 2020 AACR Virtual Meeting April 28, 2020 Symposium: New Drugs on the Horizon Part 3 12:30-1:25 PM

Reporter: Stephen J. Williams, PhD

New Drugs on the Horizon: Part 3
Introduction

Andrew J. Phillips, C4 Therapeutics

  • symposium brought by AACR CICR and had about 30 proposals for talks and chose three talks
  • unfortunately the networking event is not possible but hope to see you soon in good health

ABBV-184: A novel survivin specific T cell receptor/CD3 bispecific therapeutic that targets both solid tumor and hematological malignancies

Edward B Reilly
AbbVie Inc. @abbvie

  • T-cell receptors (TCR) can recognize the intracellular targets whereas antibodies only recognize the 25% of potential extracellular targets
  • survivin is expressed in multiple cancers and correlates with poor survival and prognosis
  • CD3 bispecific TCR to survivn (Ab to CD3 on T- cells and TCR to survivin on cancer cells presented in MHC Class A3)
  • ABBV184  effective in vivo in lung cancer models as single agent;
  • in humanized mouse tumor models CD3/survivin bispecific can recruit T cells into solid tumors; multiple immune cells CD4 and CD8 positive T cells were found to infiltrate into tumor
  • therapeutic window as measured by cytokine release assays in tumor vs. normal cells very wide (>25 fold)
  • ABBV184 does not bind platelets and has good in vivo safety profile
  • First- in human dose determination trial: used in vitro cancer cell assays to determine 1st human dose
  • looking at AML and lung cancer indications
  • phase 1 trial is underway for safety and efficacy and determine phase 2 dose
  • survivin has very few mutations so they are not worried about a changing epitope of their target TCR peptide of choice

The discovery of TNO155: A first in class SHP2 inhibitor

Matthew J. LaMarche
Novartis @Novartis

  • SHP2 is an intracellular phosphatase that is upstream of MEK ERK pathway; has an SH2 domain and PTP domain
  • knockdown of SHP2 inhibits tumor growth and colony formation in soft agar
  • 55 TKIs there are very little phosphatase inhibitors; difficult to target the active catalytic site; inhibitors can be oxidized at the active site; so they tried to target the two domains and developed an allosteric inhibitor at binding site where three domains come together and stabilize it
  • they produced a number of chemical scaffolds that would bind and stabilize this allosteric site
  • block the redox reaction by blocking the cysteine in the binding site
  • lead compound had phototoxicity; used SAR analysis to improve affinity and reduce phototox effects
  • was very difficult to balance efficacy, binding properties, and tox by adjusting stuctures
  • TNO155 is their lead into trials
  • SHP2 expressed in T cells and they find good combo with I/O with uptick of CD8 cells
  • TNO155 is very selective no SHP1 inhibition; SHP2 can autoinhibit itself when three domains come together and stabilize; no cross reactivity with other phosphatases
  • they screened 1.5 million compounds and got low hit rate so that is why they needed to chemically engineer and improve on the classes they found as near hits

Closing Remarks

 

Xiaojing Wang
Genentech, Inc. @genentech

Follow on Twitter at:

@pharma_BI

@AACR

@CureCancerNow

@pharmanews

@BiotechWorld

@HopkinsMedicine

#AACR20

Read Full Post »

Real Time Coverage @BIOConvention #BIO2019: What’s Next: The Landscape of Innovation in 2019 and Beyond. 3-4 PM June 3 Philadelphia PA

Reporter: Stephen J. Williams, PhD @StephenJWillia2

 

Results from Clarivate
In 2018 most of deals were in CART area but now we are seeing more series A rounds that are on novel mechanisms as well as rare diseases.  US is still highest in venture capital series A but next is China. 10 of top ex US VC are from China, a whole lot of money.
Preclinical is very strong for US VC but China VC is focused on clinical.  First time this year we see US series A break above 100.  But ex US the series A is going down.  Although preclinical deals in US is coming back not like as good as in 2006.  But alot of > 1 billion $ deals.  Most of money into mAbs and protein therapy;  antisense is big and cell therapy is big too; small molecule not as much
ClearView Healthcare
Which innovation classes attracted VC in 2018?
  • Oncology drives a disproportionate focus could be driven by pharma focus on oncology; however there is some focus on neuro and infectious disease
  • therapeutic classes: shift to differentiated technology…. companies want technologic platforms not just drugs.  Nucleic Acid tech and antibody tech is high need platforms.  Startups can win by developing a strong platform not just a drug
There are pros and cons of developing a platform company versus a focused company.  Many VCs have a portfolio and want something to fit in so look for a focused company and may not want a platform company.  Pfizer feels that when alot of money is available (like now) platform investing is fine but when money becomes limited they will focus on those are what will be needed to fill therapy gaps.  They believe buy the therapy and only rent the platform.
Merck does feel the way Pfizer does but they have separate ventures so they can look and license platforms.  they are active in looking at companies with new modalities but they are focused on the money so they feel best kept in hands of biotech not pharma.
At Celgene they were solely focused on approvals not platforms.  Alot of money is required to get these platforms to market.  Concentration for platform companies should be the VCs not partnering or getting bought out by pharma.  it seems from panel speakers from pharma that they are waiting for science to prove itself and waiting for favorable monetary environments (easy money).  However it seems they (big pharma) are indicating that money is drying up or at least expect it too.
At Axial and with VCs they feel it is important to paint a picture or a vision at the early stage.
At Ontogeny, they focus on evaluating assets especially and most important, ThE MANAGEMENT TEAM.  There are not that many great talented drug development management teams he feels out there even though great science out there.

Read Full Post »

Virtual Event from SelectBio: Academic Drug Discovery 2016

Reporter: Stephen J. Williams, PhD

Virtual Event from SelectBio

Academic Drug Discovery 2016

Registration to this Virtual Event is free of charge and gives you 180 days access. You will be sent details of how to access the event via email after registration.

For more information please click here.

Talks

Academic Drug Discovery: Opportunities For Challenging Targets In Oncology
Olivia Rossanese, Head of Biology and Reader, CR UK Cancer Therapeutics Unit, Institute of Cancer ResearchThe Cancer Research UK Cancer Therapeutics Unit at the Institute of Cancer Research is a multidisciplinary drug discovery group with a focus on novel, high-risk targets in cancer. This class of targets comes with unique challenges, such as a higher target validation burden or increased technical risk, but presents distinct opportunities for discovering novel biology and innovative medicines.
Academics and academic institutions need to pull together, to help generate more novel medicines for patients
Chas Bountra, Chief Scientist, University of OxfordAcademics and academic institutions need to pull together, to help generate more novel medicines for patients. Together, we are creating a new ecosystem for drug discovery. One which we believe will accelerate the generation of more novel medicines, more quickly. We hope these will also be more affordable.
Chemical Proteomics: Accelerating Academic Target Discovery And Validation
Edward Tate, Professor of Chemical Biology, Imperial College LondonMy group develops chemical biology approaches to identify and validate potential drug targets, particularly in the field of protein post-translational modification. In this talk I will present our recent research on developing chemical probes for protein lipidation, which has led to fundamental biological insights into the scope and roles of these modifications in disease biology, and to drug development projects in infection and cancer.
Institute For Applied Cancer Science, Developing The Next Generation Of Oncology Agents For Targeted Patient Populations
Philip Jones, Head of Drug Discovery, Institute for Applied Cancer Science. MD Anderson Cancer Center, University of Texas MD Anderson Cancer CenterThe Institute for Applied Cancer Science (IACS) is a fully integrated drug discovery and development unit embedded within MD Anderson. By housing an innovative drug development operation within the world’s leading cancer center, we are surrounded by the culture of innovative academic science and clinical excellence for which MD Anderson is known.
Opening “The Box of Delights” – Accessing Pharma to Enhance Academic Drug Discovery
Justin Bryans, Director of Drug Discovery, Medical Research Council TechnologyThis presentation will describe some of the ground breaking collaborations between MRC Technology and various Pharma companies, highlighting why these are so important to each party, and how these collaborations will drive innovative drug discovery and deliver new treatments to patients.
Small Molecules That Stabilize And Activate Lipoprotein Lipase (LPL)
Mikael Elofsson, Professor, University of UmeåThe presentation covers a screening-based approach to identify small molecules that stabilize and activate lipoprotein lipase, a key enzyme in lipid metabolism. A medicinal program led to improved compounds with efficacy in vivo.

Read Full Post »

Crystal Resolution in Raman Spetctoscopy for Pharmaceutical Analysis, Volume 2 (Volume Two: Latest in Genomics Methodologies for Therapeutics: Gene Editing, NGS and BioInformatics, Simulations and the Genome Ontology), Part 1: Next Generation Sequencing (NGS)

Crystal Resolution in Raman Spetctoscopy for Pharmaceutical Analysis

Curator: Larry H. Bernstein, MD, FCAP

 

Investigating Crystallinity Using Low Frequency Raman Spectroscopy: Applications in Pharmaceutical Analysis

http://images.alfresco.advanstar.com/alfresco_images/pharma/2016/01/21/52099838-6354-4ad4-b6f9-9c7c8061f307/Gordon-figure01_web.jpg

Figure 1: Illustration of an exemplar low-frequency Raman setup with a 785-nm laser.

The second system is based on a pre-built SureBlock XLF-CLM THz-Raman system from Ondax Inc. The laser (830 nm, 200 mW), cleanup filters, and laser line filters are all self-contained inside of the instrument but operate on the same principles as the 785-nm system. The sample is arranged in a 180° backscattering geometry relative to a 10× microscope lens. This system is then coupled via a fiber-optic cable to a Princeton Instruments SP2150i spectrograph and PIXIS 100 CCD camera. The 0.15-m spectrograph is used in conjunction with either a 1200- or 1800-groove/mm blazed diffraction grating to adjust the resolution and spectral range.

Crystalline Versus Amorphous Samples

The Raman spectrum of crystalline and amorphous solids differ greatly in the low-frequency region (see Figure 2) because of the highly ordered and highly disordered molecular environments of the respective solids. However, the mid-frequency region can also be noticeably altered by the changing environment (Figure 3).

 

 

http://images.alfresco.advanstar.com/alfresco_images/pharma/2016/01/21/52099838-6354-4ad4-b6f9-9c7c8061f307/Gordon-figure03_web.jpg

Figure 3: Raman spectra of griseofulvin

Ensuring Accuracy

A potential issue is optical artifacts, and these may be identified by the analysis of both Stokes and anti-Stokes spectra. One advantage of the experimental setups described is that signal from the sample may be measured within minutes and it is nondestructive, thus allowing Raman spectra to be collected from a single sample using both techniques at virtually the same time. This approach permits the examination of low-frequency Raman data with 785-nm and 830-nm excitation and allows comparison with Fourier transform (FT)-Raman spectra, in which it is possible to collect meaningful data down to a Raman shift of 50 cm-1. The benefits are demonstrated in Figure 4. In this data, each technique produces consistent bands with similar Raman shifts and relative intensities. While Raman data were not collected below 50 cm-1 using the 1064-nm system, the bands at 69 and 96 cm-1 are consistent with the 785- and 830-nm data. Furthermore, the latter two methods show consistency with bands appearing around 32 and 46 cm-1 for both techniques.

http://images.alfresco.advanstar.com/alfresco_images/pharma/2016/01/21/52099838-6354-4ad4-b6f9-9c7c8061f307/Gordon-figure04_web.jpg

Figure 4: Comparison of the low-frequency region of three Raman spectroscopic techniques.

Case Studies

So far there have been few studies to utilize low-frequency Raman spectroscopy in the analysis of pharmaceutical crystallinity. Despite this, the literature does contain articles that demonstrate the promising applicability of the technique.

Mah and colleagues (38) studied the level of crystallinity of griseofulvin using low-frequency Raman spectroscopy with PLS analysis. In this study a batch of amorphous griseofulvin (which was checked using X-ray powder diffractometry) was prepared by melting the griseofulvin and rapidly cooling it again using liquid nitrogen. Condensed water was removed by placing the sample over phosphorus pentoxide and the glassy sample was then ground using mortar and pestle. Calibrated samples of 2%, 4%, 6%, 8%, and 10% crystallinity were then created though geometric mixing of the amorphous and crystalline samples; following this mixing, the samples were then pressed into tablets. Many tablets were then stored in differing temperatures (30 °C, 35 °C, and 40 °C) at 0% humidity. Low-frequency 785-nm, mid-frequency 785-nm, and FT-Raman spectroscopies were performed simultaneously on each sample. After PLS analysis, limits of detection (LOD) and limits of quantification (LOQ) were calculated. The results of this research showed that each of these three techniques were capable of quantifying crystallinity. It also showed that FT-Raman and low-frequency Raman techniques were able to both detect and quantify crystallinity earlier than the mid-frequency 785 nm Raman technique. The respective LOD and LOQ values for FT-Raman, low-frequency Raman, and mid-frequency Raman are as follows: LOD values: 0.6%, 1.1%, and 1.5%; LOQ values: 1.8%, 3.4%, and 4.6%. The root mean squared errors of prediction (RMSEP) were also calculated and, like the LOD and LOQ values, indicated that the FT-Raman data had the lowest error, followed by the low-frequency Raman, and mid-frequency Raman had the largest errors of the three techniques. The recrystallization tests that were performed indicated that higher temperatures showed a distinct increase in the rate of recrystallization and that each technique provided similar results (within experimental error). It is also important to note that each technique gave similar spectra (where applicable), which provides supporting evidence that the data is meaningful. Overall, the conclusions of this research were that low-frequency predictions of crystallinity are at least as accurate as the predictions made using mid-frequency Raman techniques. It is arguable that low-frequency Raman is better because of the presence of stronger spectral features and because they are intrinsically linked with crystallinity.

Hédoux and colleagues (36) investigated the crystallinity of indomethacin using low-frequency Raman spectroscopy and compared the results with high frequency data. The ranges of interest were indicated to be 5–250 cm-1and 1500–1750 cm-1 regions. Samples of indomethacin were milled using a cryogenic mill to avoid mechanical heating of the sample, with full amorphous samples being obtained after 25 min of milling. Methods used in this study include Raman spectroscopy, isothermal differential scanning calorimetry (DSC), and X-ray diffractometry as well as the milling technique. The primary objective of this research was to use all of these techniques to monitor the crystallization of amorphous indomethacin to the more stable γ-state while the sample was at room temperature–well below the glass transition temperature,Tg = 43 °C. The results of this research did in fact show that low-frequency Raman spectroscopy is a very sensitive technique for identifying very small amounts of crystallinity within mostly amorphous samples. The data was supported by the well-established methods for monitoring crystallinity: XRD and DSC. This paper particularly noted the benefit of low acquisition times associated with low-frequency Raman spectroscopy compared with the other techniques used.

Low-frequency Raman spectroscopy was also used to monitor two polymorphic forms of caffeine after grinding and pressurization of the samples (39). Pressurization was performed hydrostatically using a gasketed membrane diamond anvil cell (MDAC), while ball milling was used as the method of grinding the sample. Analysis methods used were low-frequency Raman and X-ray diffraction. Low-frequency Raman spectra revealed that, upon slight pressurization, caffeine form I transforms into a metastable state slightly different from that of form II and that a disordered (amorphous) state is achieved in both forms when pressurized above 2 GPa. In contrast, it is concluded that grinding results in the transformation of each form into the other with precise grinding times, thus also generating an intermediate form, which was found to only be observable using low-frequency Raman spectroscopy. The caffeine data, as well as the low-frequency data obtained for indomethacin were further discussed by Hédoux and colleagues (40).

Larkin and colleagues (41) used low-frequency Raman in conjunction with other techniques to characterize several different APIs and their various forms. The other techniques include FT-Raman spectroscopy, X-ray powder diffraction (XRPD), and single-crystal X-ray diffractometry. The APIs studied include carbamazepine, apixaban diacid co-crystals, theophylline, and caffeine and were prepared in various ways that are not detailed here. During this research, low-frequency Raman spectroscopy played an important role in understanding the structures while in their various forms. However, more importantly, low-frequency Raman spectroscopy produced information-rich regions below 200 cm-1 for each of the crystalline samples and noticeably broad features when the APIs were in solution.

Wang and colleagues (42) investigated the applicability of low-frequency Raman spectroscopy in the analysis of respirable dosage forms of various pharmaceuticals. The analyzed pharmaceuticals were involved in the treatment of asthma or chronic obstructive pulmonary disease (COPD) and include salmeterol xinafoate, formoterol fumarate, glycopyrronium bromide, fluticasone propionate, mometasone furoate, and salbutamol sulfate. Various formulations of amino acid excipients were also analyzed in this study. Results indicated that the use of low-frequency Raman analysis was beneficial because of the large features found in the region and allowed for reliable identification of each of the dosage forms. Not only this, it also allowed unambiguous identification of two similar bronchodilators, albuterol (Ventolin) and salbutamol (Airomir).

Heyler and colleagues (43) collected both the low-frequency and fingerprint region of Raman spectra from several polymorphs of carbamazepine, an anticonvulsant and mood stabilizer. This study found that the different polymorphs of this API could be distinguished effectively using these two regions. Similarly, Al-Dulaimi and colleagues (44) demonstrated that polymorphic forms of paracetamol, flufenamic acid, and imipramine hydrochloride could be screened using low-frequency Raman and only milligram quantities of each drug. In this study, paracetamol and flufenamic acid were used as the model compounds for comparison with a previously unstudied system (imipramine hydrochloride). Features within the low-frequency Raman regions of spectra were shown to be significantly different between forms of each drug. Therefore this study also indicated that the polymorphs were highly distinguishable using the technique. Hence, like all other previously mentioned case studies, these investigations further demonstrate the utility of low-frequency Raman spectroscopy as a fast and effective method for screening pharmaceuticals for crystallinity.

Conclusions

Low-frequency Raman spectroscopy is a new technique in the field of pharmaceuticals, as well as in general studies of crystallinity. This is despite indications in previous studies showing an innate ability of the technique for identifying crystalline materials and in some cases, quantifying crystallinity. Arguably one of the most beneficial aspects of this technique is the relatively small amount of time necessary to prepare and analyze samples when compared with XRD or DSC. This should ensure the growing use of low-frequency Raman spectroscopy in, not only pharmaceutical crystallinity studies, but also crystallinity studies of other substances as well.

References

  1. J.R. Ferraro and K. Nakamoto, Introductory Raman Spectroscopy, 1st Edition (Academic Press, San Diego, 1994).
  2. K.C. Gordon and C.M. McGoverin, Int. J. Pharm. 417, 151–162 (2011).
  3. D. Law et al., J. Pharm. Sci. 90, 1015–1025 (2001).
  4. G.H. Ward and R.K. Schultz, Pharm. Res. 12, 773–779 (1995).
  5. M.D. Ticehurst et al., Int. J. Pharm. 193, 247–259 (2000).
  6. M. Rani, R. Govindarajan, R. Surana, and R. Suryanarayanan, Pharm. Res. 23, 2356–2367 (2006).
  7. M.J. Pikal, in Polymorphs of Pharmaceutical Solids, H.G. Brittain, Ed. (Marcel Dekker, New York, 1999), pp. 395–419.
  8. M. Ohta and G. Buckton, Int. J. Pharm. 289, 31–38 (2005).
  9. J. Han and R. Suryanarayanan, Pharm. Dev. Technol. 3, 587–596 (1998).
  10. S. Debnath and R. Suryanarayanan, AAPS PharmSciTech. 5, 1–11 (2004).
  11. C.J. Strachan, T. Rades, D.A. Newnham, K.C. Gordon, M. Pepper, and P.F. Taday, Chem. Phys. Lett. 390, 20–24 (2004).
  12. Y.C. Shen, Int. J. Pharm. 417, 48–60 (2011).
  13. G.W. Chantry, in Submillimeter Spectroscopy: A Guide to the Theoretical and Experimental Physics of the Far Infrared, 1st Edition (Academic Press Inc. Ltd., Waltam, 1971).
  14. D. Tuschel, Spectroscopy 30(9), 18–31 (2015).
  15. P.M.A. Sherwood, Vibrational Spectroscopy of Solids (Cambridge University Press, Cambridge, 1972).
  16. L. Ho et al., J. Control. Release. 119, 253–261 (2007).
  17. V.P. Wallace et al., Faraday Discuss. 126, 255–263 (2004).
  18. F.S. Vieira and C. Pasquini, Anal. Chem. 84, 3780–3786 (2014).
  19. J. Darkwah, G. Smith, I. Ermolina, and M. Mueller-Holtz, Int. J. Pharm.455, 357–364 (2013).
  20. S. Kojima, T. Shibata, H. Igawa, and T. Mori, IOP Conf. Ser. Mater. Sci. Eng. 54, 1–6 (2014).
  21. T. Shibata, T. Mori, and S. Kojima, Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 150, 207–211 (2015).
  22. S.P. Delaney, D. Pan, M. Galella, S.X. Yin, and T.M. Korter, Cryst. Growth Des. 12, 5017–5024 (2012).
  23. M.D. King, W.D. Buchanan, and T.M. Korter, Anal. Chem. 83, 3786–3792 (2011).
  24. C.J. Strachan et al., J. Pharm. Sci. 94, 837–846 (2005).
  25. C.M. McGoverin, T. Rades, and K.C. Gordon, J. Pharm. Sci. 97, 4598–4621 (2008).
  26. A. Heinz, C.J. Strachan, K.C. Gordon, and T. Rades, J. Pharm. Pharmacol. 971–988 (2009).,<>
  27. H.G. Brittain, J. Pharm. Sci. 86, 405–412 (1997).
  28. L. Yu, S.M. Reutzel, and G. A. Stephenson, Pharm. Sci. Technol. Today1, 118–127 (1998).
  29. M. Dracínský, E. Procházková, J. Kessler, J. Šebestík, P. Matejka, and P. Bour, J. Phys. Chem. B. 117, 7297–7307 (2013).
  30. P. Sharma et al., J. Raman Spectrosc. DOI 10.1002/jrs.4834, wileyonlinelibrary.com (2015).
  31. A.P. Ayala, Vib. Spectrosc. 45, 112–116 (2007).
  32. J.F. Scott, Spex Speak. 17, 1–12 (1972).
  33. D.P. Strommen and K. Nakamoto, in Laboratory Raman Spectroscopy, 1st Edition (John Wiley & Sons Inc., New York, 1984).
  34. A.L. Glebov, O. Mokhun, A. Rapaport, S. Vergnole, V. Smirnov, and L.B. Glebov, Proc. SPIE. 8428, 84280C1–84280C11 (2012).
  35. E.P.J. Parrott and J.A. Zeitler, Appl. Spectrosc. 69, 1–25 (2015).
  36. A. Hédoux, L. Paccou, Y. Guinet, J.F. Willart, and M. Descamps, Eur. J. Pharm. Sci. 38, 156–164 (2009).
  37. R.L. McCreery, in Raman Spectroscopy for Chemical Analysis, 1st Edition (John Wiley & Sons Inc., New York, 2000).
  38. P.T. Mah, S.J. Fraser, M.E. Reish, T. Rades, K.C. Gordon, and C.J. Strachan, Vib. Spectrosc. 77, 10–16 (2015).
  39. A. Hédoux, A.A. Decroix, Y. Guinet, L. Paccou, P. Derollez, and M. Descamps, J. Phys. Chem. B. 115, 5746–5753 (2011).
  40. A. Hédoux, Y. Guinet, and M. Descamps, Int. J. Pharm. 417, 17–31 (2011).
  41. P.J. Larkin, M. Dabros, B. Sarsfield, E. Chan, J.T. Carriere, and B.C. Smith, Appl. Spectrosc. 68, 758–776 (2014).
  42. H. Wang, M. A. Boraey, L. Williams, D. Lechuga-Ballesteros, and R. Vehring, Int. J. Pharm. 469, 197–205 (2014).
  43. R. Heyler, J. Carriere, and B. Smith, in “Raman Technology for Today’s Spectroscopists,” supplement to Spectroscopy (June), 44–50 (2013).
  44. S. Al-Dulaimi, A. Aina, and J. Burley, CrystEngComm. 12, 1038–1040 (2010).

 

The drawing in Figure 1 is that of a six-membered ring or hexagon. A carbon atom is located at each vertex of the hexagon and a hydrogen atom is attached to each carbon, although it is not written in. The circle inside the ring represents that the electrons are delocalized which is illustrated in Figure 2.

http://images.alfresco.advanstar.com/alfresco_images/pharma/2016/02/12/645ee751-2432-4444-8af1-ded62697ee27/IR-Spectral-figure02_web.jpg

Figure 2: Top: The P orbitals on each of the six carbon atoms in benzene that contribute an electron to the ring. Bottom: the collection of delocalized P orbital electrons forming a cloud of electron density above and below the benzene ring.

Each of the carbon atoms in a benzene ring contains two P orbitals containing a lone electron, and one of these orbitals is perpendicular to the benzene ring as seen in the top of Figure 2. There is enough orbital overlap that these electrons, rather than being confined between two carbon atoms as might be expected, instead delocalize and form clouds of electron density above and below the plane of the ring. This type of bonding is called aromatic bonding(2), and a ring that has aromatic bonding is called an aromatic ring. It is aromatic bonding that gives aromatic rings their unique structures, chemistry, and IR spectra. Benzene is simply a commonly found aromatic ring. Other types of aromatic molecules include polycyclic aromatic hydrocarbons (PAHs), such as naphthalene, that contain two or more benzene rings that are fused (which means adjacent rings share two carbon atoms), and heterocyclic aromatic rings which are aromatic rings that contain a noncarbon atom such as nitrogen. Pyridine is an example of one of these. The interpretation of the IR spectra of these latter aromatic molecules will be discussed in future articles.

The IR Spectrum of Benzene

The IR spectrum of benzene is shown in Figure 3.

http://images.alfresco.advanstar.com/alfresco_images/pharma/2016/02/12/645ee751-2432-4444-8af1-ded62697ee27/IR-Spectral-figure03_web.jpg

 

09:00

Super-Resolution Fluorescence Microscopy: Where To Go Now?
Bernd Rieger, Quantitative Imaging Group Leader, Delft University of Technology

09:30

Keynote Presentation

From Molecules To Whole Organs
Francesco Pavone, Principal Investigator, LENS, University of Florence

Some examples of correlative microscopies, combining linear and non linear techniques will be described. Particular attention will be devoted Alzheimer disease or to neural plasticity after damage as neurobiological application.

10:15

Super-Resolution Imaging by dSTORM
Markus Sauer, Professor, Julius-Maximilians-Universität Würzburg

10:45

Coffee and Networking in Exhibition Hall

11:15

Correlated Fluorescence And X-Ray Tomography: Finding Molecules In Cellular CT Scans
Carolyn Larabell, Professor, University of California San Francisco

11:45

Integrating Advanced Fluorescence Microscopy Techniques Reveals Nanoscale Architecture And Mesoscale Dynamics Of Cytoskeletal Structures Promoting Cell Migration And Invasion
Alessandra Cambi, Assistant Professor, University of Nijmegen

This lecture will describe our efforts to exploit and integrate a variety of advanced microscopy techniques to unravel the nanoscale structural and dynamic complexity of individual podosomes as well as formation, architecture and function of mesoscale podosome clusters.

12:15

Multi-Photon-Like Fluorescence Microscopy Using Two-Step Imaging Probes
George Patterson, Investigator, National Institutes of Health

12:45

Lunch & Networking in Exhibition Hall

14:15

Technology Spotlight

14:30

3D Single Particle Tracking: Following Mitochondria in Zebrafish Embryos
Don Lamb, Professor, Ludwig-Maximilians-University

15:00

Visualizing Mechano-Biology: Quantitative Bioimaging Tools To Study The Impact Of Mechanical Stress On Cell Adhesion And Signalling
Bernhard Wehrle-Haller, Group Leader, University of Geneva

15:30

Superresolution Imaging Of Clathrin-Mediated Endocytosis In Yeast
Jonas Ries, Group Leader, EMBL Heidelberg

We use single-molecule localization microscopy to investigate the dynamic structural organization of the east endocytic machinery. We discovered a striking ring-shaped pre-patterning of the actin nucleation zone, which is key for an efficient force generation and membrane invagination.

16:00

Coffee and Networking in Exhibition Hall

16:30

Optical Imaging of Molecular Mechanisms of Disease
Clemens Kaminski, Professor, University of Cambridge

17:00

3-D Optical Tomography For Ex Vivo And In Vivo Imaging
James McGinty, Professor, Imperial College London

17:30

End Of Day One

Wednesday, 15 June 2016

09:00

Imaging Gene Regulation in Living Cells at the Single Molecule Level
James Zhe Liu, Group Leader, Janelia Research Campus, Howard Hughes Medical Institute

09:30

Keynote Presentation

Super-Resolution Microscopy With DNA Molecules
Ralf Jungmann, Group Leader, Max Planck Institute of Biochemistry

10:15

A Revolutionary Miniaturised Instrument For Single-Molecule Localization Microscopy And FRET
Achillefs Kapanidis, Professor, University of Oxford

10:45

Coffee and Networking in Exhibition Hall

11:15

Democratising Live-Cell High-Speed Super-Resolution Microscopy
Ricardo Henriques, Group Leader, University College London

11:45

Democratising Live-Cell High-Speed Super-Resolution Microscopy

12:15

Information In Localisation Microscopy
Susan Cox, Professor, Kings College London

12:45

Lunch & Networking in Exhibition Hall

14:15

Technology Spotlight

14:30

High-Content Imaging Approaches For Drug Discovery For Neglected Tropical Diseases
Manu De Rycker, Team Leader, University of Dundee

The development of new drugs for intracellular parasitic diseases is hampered by difficulties in developing relevant high-throughput cell-based assays. Here we present how we have used image-based high-content screening approaches to address some of these issues.

15:00

High Resolution In Vivo Histology: Clinical in vivo Subcellular Imaging using Femtoseceond Laser Multiphoton/CARS Tomography
Karsten König, Professor, Saarland University

We report on a certified, medical, transportable multipurpose nonlinear microscopic imagingsystem based on a femtosecond excitation source and a photonic crystal fiber with multiple miniaturized time-correlated single-photon counting detectors.

15:30

Coffee and Networking in Exhibition Hall

16:00

Lateral Organization Of Plasma Membrane Constituents At The Nanoscale
Gerhard Schutz, Professor, Vienna University of Technology

It is of interest how proteins are spatially distributed over the membrane, and whether they conjoin and move as part of multi-molecular complexes. In my lecture, I will discuss methods for approaching the two questions, and provide biological examples.

16:30

Correlative Light And Electron Microscopy In Structural Cell Biology
Wanda Kukulski, Group Leader, University of Cambridge

17:00

Close of Conference

 

Read Full Post »

Mid Atlantic LRIG 22nd Annual Technology Showcase: Agenda on 3D Bioprinting on Wednesday, May 11, 2016 at Holiday Inn, 195 Davidson Avenue, Somerset, NJ

Reporter: Stephen J. Williams, Ph.D.

 

Symposium Speakers and Topics:

Human Organoids
Hatem E. Sabaawy-Director, Production GMP Facility for Cell and Gene Therapy, RBHS-Robert Wood Johnson Medical School, Rutgers Cancer Institute of New Jersey

Intestinal Organoids for Drug Discovery
Richard Visconti-Associate Principal Scientist, Cellular Pharmacology, Merck Research Laboratories, Kenilworth,  New Jersey

3D Bioprinting
Elizabeth Wu-President, WuZenTech, Edison, New Jersey

Building  Your Brand  Through LinkedIn
Stan Robinson, Jr., LinkedIn Consultant, Helping Professionals with Social Selling, Personal Branding

Register at EventBrite here: https://www.eventbrite.com/e/mid-atlantic-22nd-annual-technology-and-exhibition-tickets-21359945171 

To sign up to be an LRIG member or update your profile, please visit us at http://lrig.org
Hoping to see you on May 11th.
Reserve your spot today!

 

Read Full Post »

2nd Annual Translational Gene Editing: Exploiting CRISPR/Cas9 for Building Tools for Drug Discovery & Development: June 16, 2016, Boston, MA

Reporter: Stephen J. Williams, PhD

Translational Gene Editing – June 16-17, 2016 in Boston, MA

YouTubeLinkedInTwitter#CHIWPC16

Learn More | Sponsorship & Exhibit Details | Register by April 29 & SAVE up to $200!

IMPROVING CRISPR FOR BETTER FUNCTIONAL SCREENING

Optimized sgRNA Libraries for Genetic Screens with CRISPR-Cas9
John Doench, Ph.D., Associate Director, Genetic Perturbation Platform, Broad Institute of Harvard and MIT

Optimizing CRISPR for Pooled Genome-Wide Functional Genetic Screens
Paul Diehl, Ph.D., Director, Business Development, Cellecta, Inc.

CRISPR-Cas9 Whole Genome Screening: Going Where No Screen Has Gone Before
Ralph Garippa, Ph.D., Director, RNAi Core Facility, Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center

Cross-Species Synthetic Lethal Screens and Applications to Drug Discovery
Norbert Perrimon, Ph.D., Professor, Department of Genetics, Harvard Medical School and Investigator, Howard Hughes Medical Institute

Interactive Breakout Discussion Groups with Continental Breakfast
This session features various discussion groups that are led by a moderator/s who ensures focused conversations around the key issues listed. Attendees choose to join a specific group and the small, informal setting facilitates sharing of ideas and active networking. Continental breakfast is available for all participants.

Topic: CRISPR/Cas9 System for In vivo Drug Discovery
Moderator: Danilo Maddalo, Ph.D., Lab Head, ONC Pharmacology, Novartis Institutes for BioMedical Research

  • Impact of CRISPR/Cas9 system on in vivo mouse models
  • Application of the CRISPR/Cas9 system in in vivo screens
  • Technical limitations/safety issues

Topic: Getting Past CRISPR Pain Points
Moderators: John Doench, Ph.D., Associate Director, Genetic Perturbation Platform, Broad Institute of Harvard and MITStephanie Mohr, Ph.D., Lecturer, Genetics & Director of the Drosophila RNAi Screening Center, Harvard Medical School

  • Challenges and solutions for CRISPR gRNA design
  • Methods for detecting engineered changes

Topic: Cellular Delivery of CRISPR/Cas9
Moderator: Daniel E Bauer M.D., Ph.D., Assistant Professor of Pediatrics, Harvard Medical School and Staff Physician in Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Principal Faculty, Harvard Stem Cell Institute

GENE EDITING FOR SCREENING DISEASE PATHWAYS AND DRUG TARGETS

Scouring the Non-Coding Genome by Saturating Edits
Daniel E. Bauer, M.D., Ph.D., Assistant Professor of Pediatrics, Harvard Medical School and Staff Physician in Pediatric Hematology/Oncology, Boston Children’s Hospital and Dana-Farber Cancer Institute, Principal Faculty, Harvard Stem Cell Institute

Parallel shRNA and CRISPR/Cas9 Screens Reveal Biology of Stress Pathways and Identify Novel Drug Targets
Michael Bassik, Ph.D., Assistant Professor, Department of Genetics, Stanford University

BUILDING THE CRISPR TOOLBOX

Beyond Cas9: Discovering Single Effector CRISPR Tools
Jonathan Gootenberg, Member, Laboratories of Dr. Aviv Regev and Dr. Feng Zhang, Department of Systems Biology, Harvard Medical School, and Broad Institute of Harvard and MIT

CRISPR-Cas9 Genome Editing Improves Sub-Cellular Localization Studies
Netanya Y. Spencer, M.D., Ph.D., Research Fellow in Medicine, Joslin Diabetes Center, Harvard Medical School

TECHNOLOGY PANEL: Trends in CRISPR Technologies
Panelists to be Announced

This panel will bring together 2-3 technical experts from leading technology and service companies to discuss trends and improvements in CRISPR libraries, reagents and platforms that users can expect to see in the near future. (Opportunities Available for Sponsoring Panelists)

APPLICATIONS OF CRISPR FOR DRUG DISCOVERY

Use of CRISPR and Other Genomic Technologies to Advance Drug Discovery
Namjin Chung, Ph.D., Head, Functional Genomics Platform, Discovery Research, AbbVie, Inc.

Application of Genome Editing Tools to Model Human Genetics Findings in Drug Discovery
Myung Shin, Ph.D., Senior Principal Scientist, Genetics and Pharmacogenomics, Merck & Co. Inc.

In vivo Application of the CRISPR/Cas9 Technology for Translational Research
Danilo Maddalo, Ph.D., Lab Head, ONC Pharmacology, Novartis Institutes for BioMedical Research

DEVELOPING TOOLS FOR BETTER TRANSLATION

Improving CRISPR-Cas9 Precision through Tethered DNA-Binding Domains
Scot A. Wolfe, Ph.D., Associate Professor, Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School

Nucleic Acid Delivery Systems for RNA Therapy and Gene Editing
Daniel G. Anderson, Ph.D., Professor, Department of Chemical Engineering, Institute for Medical Engineering & Science, Harvard-MIT Division of Health Sciences & Technology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology

Translating CRISPR/Cas9 into Novel Medicines
Alexandra Glucksmann, Ph.D., COO, Editas Medicine

2nd Annual Translational Gene Editing: Exploiting CRISPR/Cas9 for Building Tools for Drug Discovery & Development: June 16, 2016, Boston, MA, Volume 2 (Volume Two: Latest in Genomics Methodologies for Therapeutics: Gene Editing, NGS and BioInformatics, Simulations and the Genome Ontology), Part 2: CRISPR for Gene Editing and DNA Repair

Read Full Post »

GEN Tech Focus: Rethinking Gene Expression Analysis, Volume 2 (Volume Two: Latest in Genomics Methodologies for Therapeutics: Gene Editing, NGS and BioInformatics, Simulations and the Genome Ontology), Part 1: Next Generation Sequencing (NGS)

GEN Tech Focus: Rethinking Gene Expression Analysis

Larry H. Bernstein, MD, FCAP, Curator

LPBI

 

Quantitating gene expression is essential for researchers to answer important biological questions about basic cellular functions, as well as disease states. In the following articles you will discover the multitude of advances investigators have made to accurately measure and quantitate genetic transcripts within the cell.

Diverse Pathways to Drug Targets

A great deal of research on pathway analysis is currently focusing on RNA rather than proteins, and the complex RNA networks that regulate gene expression. With the realization that more than 90% of the genome that is transcribed into RNA is not translated into protein, and the growing numbers of naturally occurring microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) being identified and characterized, the important role these RNAs play in normal biological processes and across human diseases is becoming increasingly clear.

 

The Gene-Expression Undergrowth Have Been Well Trodden, but RNA Paths Want Wear, Too

  • Hepatitis C virus depends on a functional interaction between its genome and miR-122 for viral stability and replication. Researchers recently used an antisense oligonucleotide that targets the liver-specific microRNA miR-122, blocking its function. [Bluebay2014/Fotolia]

    A great deal of research on pathway analysis is currently focusing on RNA rather than proteins, and the complex RNA networks that regulate gene expression.

    With the realization that more than 90% of the genome that is transcribed into RNA is not translated into protein, and the growing numbers of naturally occurring microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) being identified and characterized, the important role these RNAs play in normal biological processes and across human diseases is becoming increasingly clear.

    This knowledge—combined with the available technology and strategies to decipher RNA pathways and link alterations in the levels or activity of miRNAs or lncRNAs to gene expression, epigenetic mechanisms, and protein activity in normal and disease phenotypes—is driving the development and clinical testing of novel drug targets and therapeutics that target regulatory RNAs.

    For example, a microRNA was targeted in a Phase II clinical study that assessed the effect of miravirsen, an antisense oligonucleotide, in patients with hepatitis C. The study, which was described in 2013 in the New England Journal of Medicine, indicated that miravirsen sequesters the liver-specific microRNA miR-122 in a highly stable heteroduplex, thereby inhibiting its function.

    Hepatitis C virus (HCV) depends on a functional interaction between its genome and miR-122 for viral stability and replication. According to the study, inhibition of miR-122 in HCV-infected patients was associated with decreased levels of HCV RNA that continued beyond the treatment period, without evidence of viral resistance.

    The therapeutic potential of regulatory RNAs is also being assessed in other conditions such as cancer. Specifically, miRNAs and other ncRNAs in cancer initiation, progression, and metastasis are being studied by George Calin, M.D., Ph.D., a professor of experimental therapeutics, MD Anderson Cancer Center, University of Texas. Dr. Calin’s group is scouring the “microRNAome” to identify miRNAs of about 21–22 nucleotides that can serve as reliable biomarkers for cancer diagnosis and to guide decision-making in patient management, including as predictors of survival and response to drug therapy.

    miRNAs are involved in every aspect of tumorigenesis, cancer progression, and dissemination. Not only are they expressed in tumor cells, they are also stably expressed in exosomes and are present in various bodily fluids, where they can act like hormones and signaling molecules. Comparative profiling of these fluids for differences in miRNA levels between patients with and without cancer could identify relevant biomarkers.

     

    Analyzing RNA Pathways

  • Using Qiagen’s Ingenuity Pathway Analysis, researchers can analyze relationships between molecules and diseases of interest by modeling how gene expression patterns affect functional outcomes or disease processes.

     

     

     

     

     

     

     

     

     

    Dr. Calin and colleagues have described the significance of miRNA signatures obtained in recent studies involving miRNA profiling of human tumors. An overview appeared 2014 in CA: A Cancer Journal for Clinicians (“MicroRNAome genome: a treasure for cancer diagnosis and therapy”). Also, last February, Dr. Calin gave an account of his group’s work at the Molecular Med Tri Conference in San Francisco.

    Technology is not holding back advances in the field of RNA pathway analysis according to Dr. Calin. The main bottleneck at present is in the design of prospective studies needed to confirm the predictive value of miRNA-based biomarkers.

    Dr. Calin points to two other key challenges that scientists currently face in translating research findings into diagnostic, prognostic, and therapeutic tools. One is the difficulty in selecting an miRNA target, mainly because an individual miRNA could have a role in regulating tens, hundreds, or even thousands of protein-coding genes. For drug discovery, the aim is to identify miRNAs that affect a single pathway of interest to help limit off-target effects. The need for novel delivery systems for RNA-targeted drugs is another key challenge.

    At the Molecular Med Tri Conference, Jean-Noel Billaud, Ph.D., principal scientist at Qiagen Bioinformatics, presented a case study demonstrating how the company’s Ingenuity Pathway Analysis technology can be used to conduct a systems biology analysis to identify the pathways, potential upstream regulators, and downstream outcomes involved in the host response to West Nile Virus (WNV) infection. Dr. Billaud also discussed how to interpret the results from a biological perspective.

    In his presentation, Dr. Billaud described the first step in this analytical process as the acquisition of RNA sequence data using next-generation sequencing techniques for the purpose of characterizing and quantifying differential gene expression between an infected and uninfected cell. The CLC Cancer Research Workbench tool is used to process the sequence data, and the results are imported directly into the IPA system.

    Analysis of differential gene expression aims to answer a series of key questions, including the following: What metabolic and/or signaling pathway(s) is activated or inhibited? Is there an overlap of the genes or pathways that are activated or inhibited? What are the potential upstream, downstream, functional, and phenotypic implications of this pathway activation or inhibition?

    Dr. Billaud described other questions researchers might attempt to answer through the use of IPA: What are the identifying the underlying transcriptional programs? Which biological processes are involved and in what way? Are there splice variants of interest? What type of regulation is involved?

    In the WNV case study, IPA predicted activation of the interferon signaling pathway and added statistically and functionally relevant biological processes to the WNV-related biochemical network the system developed. IPA is able to simulate the effects of interferon pathway activation on neighboring molecules and processes, which enables broader modeling of antiviral responses, prediction of the effects on viral replication, and identification of upstream transcriptional regulators of antiviral and related anti-inflammatory processes, for example.

    These data and analytical capabilities may allow researchers to propose new hypotheses that connect molecules in regulatory networks to disease-related pathways in a predictive way, leading to the identification of a “master regulator” that could serve as a disease-specific drug target, according to Dr. Billaud.

    In the WNV example, he described the use of the Molecule Activity Predictor (MAP) function in IPA to test the hypothesis that CLEC7A is a host susceptibility factor required by WNV to stimulate an immune response in the brains of infected patients, contributing to the development of life-threatening encephalitis. The MAP function simulates the inhibition or downregulation of CLEC7A, showing how it would likely reduce the risk of WNV-associated encephalitis. These types of hypotheses would then need to be tested and validated.

    Pathways Driving B-Cell Differentiation

    • Robert C. Rickert, Ph.D., professor and director of the Tumor Microenvironment and Metastasis Program at Sanford-Burnham Medical Research Institute, is using conditional gene targeting to identify the genes and biochemical pathways that play a role at specific stages of B-cell differentiation. With this approach, it is possible to knock out targeted genes in a mouse at different stages of B-cell development, and to do so in an inducible fashion, allowing you “to look at how it affects different signal transduction pathways in a context-specific manner,” says Dr. Rickert.

      When applied to a relevant mouse model of disease—such as a B-cell lymphoma—this inducible genetic system should yield effects similar to those that could be obtained with a drug capable of blocking the activity of the targeted gene product. Dr. Rickert and colleagues are exploring the similarity between the effects achieved with conditional gene targeting and those of recently approved drugs to treat chronic lymphocytic leukemia (CLL) and some forms of lymphoma such as idelalisib and ibrutinib, which are both inhibitors of the B-cell receptor pathway via blocking of PI3K or Bruton’s tyrosine kinase (BTK), respectively.

      Dr. Rickert presented his group’s latest research at a Keystone Symposium Conference, PI 3-Kinase Signaling Pathways in Disease, which took place last January in Vancouver. In his talk, Dr. Rickert emphasized that the phosphatidyl inositol-3 kinase (PI3K) pathway is a major regulator B lymphocyte differentiation and function.

      Dr. Rickert has also applied conditional gene targeting to compare the roles of the NFκB and PI3K pathways in B-cell maturation. He has shown that while both pathways are essential at some stages of B-cell differentiation, only one pathway may be necessary for B-cell maintenance and survival.

      “Ultimately we want to gain more insight at the biochemical level into single cells and the heterogeneity of the cell populations we’re interested in,” says Dr. Rickert. Tumors and cancer cell populations are quite heterogeneic, and better biochemical tools are needed to be able to sort through these populations of cells and “look at some of the more interesting, rogue cells, such as cancer stem cells,” he adds.

    An Evolutionary Approach

    In his laboratory at Hebrew University of Jerusalem, researcher Yuval Tabach, Ph.D., is using computational tools to analyze and compare the genomes and proteins of hundreds of species to identify evolutionary patterns of conservation and loss that point to connections between molecular pathways and disease.

    “The main power of this phylogenetic profiling approach is that if you look at proteins across evolution, some are lost at certain points in certain species,” says Dr. Tabach. For example, proteins involved in the tricarboxylic acid (TCA) cycle have been highly conserved across some species, but have disappeared in others because those species have lost their mitochondria.

    Dr. Tabach and colleagues have shown that sets of genes associated with particular diseases have similar phylogenetic profiles. They are also using this approach to identify genes associated with longevity, cancer resistance, and various extreme environmental conditions.

    Phylogenetic profiling to connect patterns of conservation and loss across millions of years of evolution can be applied to entire proteins, protein domains, and RNA molecules such as microRNAs. The potential applicability of this approach to drug discovery and development is multifaceted.

    For example, given a gene known to be related to a certain disease, the ability to identify other genes with a similar phylogenetic profile might reveal genetic factors that could explain incomplete penetrance or the variability of disease severity in different affected individuals. Alternatively, identification of a candidate gene in one patient could serve as the basis for identifying other key factors in other patients with the same disease using the phylogenetic profile.

    Compared to strategies such as gene expression analysis or protein-protein interaction mapping for identifying disease-related genes, phylogenetic profiling “is much faster” and will become an increasingly powerful tool as the genome sequences of more species become available, explains Dr. Tabach.

    The Israeli start-up company ReThink Pharmaceuticals is using the molecular networks generated through this phylogenetic profiling work for the purpose of drug repositioning. “If you know that a certain drug targets a gene, we can build a network to find other genes/proteins that interact with the drug target,” asserts Dr. Tabach, citing preliminary results that demonstrate the ability to predict additional effects of a drug candidate.

     

 

Measuring siRNA-mediated Knockdown of the IL-8 gene Using the QuantiGene Singleplex Assay

A critical component of RNA interference (RNAi) studies is the validation of gene expression inhibition. RNAi experiments have many sources of variation that make accurate quantitation of target mRNA difficult when qPCR is used. Variation in the potency and stability of short interfering RNA (siRNA), coupled with differences in transfection efficiency and protein turnover, results in varying gene knockdown efficiency.

 

The RNA World Expands

Over the past 10 years, scientists say new methods, including deep sequencing and DNA tiling arrays, have enabled the identification and characterization of the human transcriptome. These techniques completely changed our understanding of genome organization and content and revealed that a much larger part of the human genome is transcribed into RNA than was previously assumed—about 70%.

The RNA World Expands  

Long noncoding RNAs mean more than HOTAIR.

The RNA World Expands

Long noncoding RNA (lncRNAs) can regulate gene expression at epigenetic, transcriptional, and post-transcriptional levels. [© Alila Medicinal Media – Fotolia.com]

  • Over the past 10 years, scientists say new methods, including deep sequencing and DNA tiling arrays, have enabled the identification and characterization of the human transcriptome. These techniques completely changed our understanding of genome organization and content and revealed that a much larger part of the human genome is transcribed into RNA than was previously assumed—about 70%.

    Last year researchers, including Tim Mercer, Ph.D., at the Institute for Molecular Bioscience-University of Queensland, Roche Nimblegen, and John Rinn, Ph.D., and his team in the department of stem cell and regenerative biology at Harvard, reported that “transcriptomic analyses have revealed an ‘unexpected complexity’ to the human transcriptome, the depth and breadth of which exceeds current RNA sequencing capability.”

    These scientists used these techniques to identify and characterize unannotated transcripts whose rare or transient expression is below the detection limits of conventional sequencing approaches. The data also show that intermittent sequenced reads observed in conventional RNA sequencing datasets, previously dismissed as noise, are indicative of unassembled rare transcripts. Collectively, they say these results reveal the range, depth, and complexity of a human transcriptome that is far from fully characterized.

    Noncoding transcripts are RNA molecules that include classical “housekeeping” RNAs such as transfer RNAs (tRNAs), ribosomal RNAs (rRNAs), small nuclear RNAs (snRNAs), and small nucleolar RNAs (snoRNAs), which are constitutively expressed and play critical roles in protein biosynthesis.

    Among these noncoding RNAs are numerous long noncoding RNAs (lncRNAs), which are defined as endogenous cellular RNAs of more than 200 nucleotides in length that lack an open reading frame of significant length (less than 100 amino acids). The RNA molecules constitute a heterogeneous group, allowing them, scientists point out, to cover a broad spectrum of molecular and cellular functions by implementing different modes of action. lncRNAs are roughly classified based on their position relative to protein-coding genes as intergenic (between genes), intragenic/intronic (within genes), and antisense. Initial efforts to characterize these molecules demonstrated that they function in cis, regulating their immediate genomic neighbors.

    Regulatory Levels

  • lncRNAs can regulate gene expression at epigenetic, transcriptional, and post-transcriptional levels and take part in various physiological and pathological processes, such as cell development, immunity, oncogenesis, clinical disease processes, and more. A classic lncRNA, HOTAIR, was originally identified through work done by Howard Chang, M.D., Ph.D., at Stanford, and Dr. Rinn. Their research eventually led to the discovery of this 2.2 kilobase spliced RNA transcript that interacts with Polycomb group proteins to modify chromatin and repress transcription of the human HOX genes, which regulate development. It remains unclear as to exactly this is accomplished.

    HOTAIR, it was found, originates from the HOXC locus and represses transcription across 40 kb of that locus by altering the chromatin trimethylation state. Hox genes, a highly conserved subgroup of the homeobox superfamily, regulate numerous processes including apoptosis, receptor signaling, differentiation, motility, and angiogenesis. Aberrations in Hox gene expression have been reported in abnormal development and malignancy.

    HOTAIR works to repress Hox gene expression by directing the action of Polycomb chromatin remodeling complexes in trans to govern the cells’ epigenetic state and subsequent gene expression.HOTAIR expression is increased in primary breast tumors and metastases and its expression level in primary tumors can predict eventual metastasis and death. The recent discovery that lncRNA HOTAIRcan link chromatin changes to cancer metastasis furthers the relevance of lncRNAs to human disease.

    Dr. Chang and his colleagues say that the finding that several lncRNAs can control transcriptional alteration implies that the difference in lncRNA profiling between normal and cancer cells is not merely the secondary effect of cancer transformation, and that lncRNAs are strongly associated with cancer progression. The researchers showed that lncRNAs in the HOX loci become systematically dysregulated during breast cancer progression.

    They further demonstrated that enforced expression of HOTAIR in epithelial cancer cells induced genome-wide retargeting of polycomb repressive complex 2 (PRC2) to an occupancy pattern more resembling embryonic fibroblasts, leading to altered histone H3 lysine 27 methylation, gene expression, and increased cancer invasiveness and metastasis in a manner dependent on PRC2.

    On the other hand they noted loss of HOTAIR can inhibit cancer invasiveness, particularly in cells that possess excessive PRC2 activity. These findings indicate that lncRNAs have active roles in modulating the cancer epigenome and may be important targets for cancer diagnosis and therapy. Thus, the investigators say, differential expression of lncRNAs may be profiled to aid in cancer diagnosis and prognosis and in the selection of potential therapeutics.

    Two years ago the GENCODE consortium, within the framework of the ENCODE project, presented, and analyzed the most complete human lncRNA annotation to date. The data comprise 9,277 manually annotated genes producing 14,880 transcripts. The identification and annotation of this wealth of lncRNAs leaves scientists with a lot of research to do to fully characterize the varied functions of these unusual RNAs. Their identification also challenges technology developers to produce the tools to necessary for these analyses.

     

Transcript Regulation of 18 ADME Genes by Prototypical Inducers in Human Hepatocytes

Drug-drug interactions (DDIs) are of particular concern for regulatory agencies and the pharmaceutical industry for drug safety. Induction of drug metabolizing enzymes by pharmaceuticals, nutraceuticals, and lifestyle influences is one type of DDI in which the influence of a perpetrator molecule increases the enzyme capacity that can metabolize a victim molecule, rendering it ineffective as a therapy. To evaluate this potential, screening assays have been developed, such as the use…

 

Biomarkers Reshape Drug Development

Biomarkers defining specific phenotypes are becoming increasingly important for developing new drugs for specific patient subpopulations. The value of a new biomarker is measured by its ability to reduce risk. Ideally, the biomarker should be developed in parallel with the new drug, as nearly 50% of the projected development costs can be saved by…

Biomarkers Reshape Drug Development

  • Imanova takes a structured approach to the development of imaging biomarkers, or i-biomarkers.

    Biomarkers defining specific phenotypes are becoming increasingly important for developing new drugs for specific patient subpopulations. The value of a new biomarker is measured by its ability to reduce risk.

    Ideally, the biomarker should be developed in parallel with the new drug, as nearly 50% of the projected development costs can be saved by shutting down a development program before it enters Phase II. A meaningful risk-benefit analysis of a biomarker requires estimates of its cost and accuracy, as well as the consequences of decisions that it will enable.
    For the biomarker to be of value, the cost of its development has to be less than the projected costs of development from Phase II onwards, discounted to present time. While multiple competing business considerations affect a pharmaceutical company’s decision to proceed with a biomarker program, the skyrocketing market for biomarker discovery underscores the pharmaceutical industry’s hope that biomarkers will bolster the success rates of pipeline products.
    “Imaging biomarkers have been Ideally, the biomarker should be developed in parallel with the new drug, as nearly 50% of the projected development costs can be saved by shutting down a development program before it enters Phase II. A meaningful risk-benefit analysis of a biomarker requires estimates of its cost and accuracy, as well as the consequences of decisions that it will enable.

    Ideally, the biomarker should be developed in parallel with the new drug, as nearly 50% of the projected development costs can be saved by shutting down a development program before it enters Phase II. A meaningful risk-benefit analysis of a biomarker requires estimates of its cost and accuracy, as well as the consequences of decisions that it will enable.

    For the biomarker to be of value, the cost of its development has to be less than the projected costs of development from Phase II onwards, discounted to present time. While multiple competing business considerations affect a pharmaceutical company’s decision to proceed with a biomarker program, the skyrocketing market for biomarker discovery underscores the pharmaceutical industry’s hope that biomarkers will bolster the success rates of pipeline products.

    “Imaging biomarkers have been largely underutilized in drug development,” says Kevin Cox, Ph.D., CEO of London-based Imanova. “But we believe that molecular imaging has the power to assist in successful translation of molecules by reducing the risk of several specific causes of failure in Phase II clinical studies. Imaging biomarkers, or i-biomarkers, are especially valuable in giving confidence of tissue delivery, determination of target engagement, and the evaluation of a drug’s pharmacodynamic effects.”

    While imaging is routinely used in clinical diagnostics for cancer, its acceptance in drug development has been slow. “This is a highly specialized area of knowledge,” Dr. Cox observes. “Designing imaging experiments to answer the right questions is not trivial. Combined with the perceived high costs and dearth of well-equipped facilities, this has slowed down the adoption of imaging as an integral step in drug development.”

    Imanova presents an innovative and highly integrated solution in reducing the barriers for use of molecular imaging. Located in the former GlaxoSmithKline imaging center, Imanova’s staff applies the knowledge needed for translational application of imaging science.

    “Another historical barrier for use of molecular imaging has been the lack of versatile PET tracers for key therapeutic targets,” remarks Dr. Cox. Together with its pharmaceutical clients, Imanova develops proprietary tracers that can answer critical questions about target engagement directly after drug administration. A structured approach for i-biomarker development takes the novel tracer from the candidate pool to clinical validation.

    Uniquely, Imanova utilizes in silico biomathematical modeling to predict a candidate with ideal physicohemical characteristics. “The i-biomarker development pipeline adheres to a strict quality system,” continues Dr. Cox. “We not only provide candidate selection and labeling, but also rigorous preclinical evaluation in several species, combined with blood chemistry or other physiological measurements.”

    The resulting biomarker provides quantitative information to make informed go/no-go decisions. Imanova hopes to develop an open innovation approach to i-biomarker research, and to encourage pharmaceutical companies to collaborate on tracer development.

    “By collaborating in this pre-competitive space, a pharma-academic consortium can de-risk i-biomarker development programs and generate new tools to eliminate costs associated with futile activities downstream,” concludes Dr. Cox. “Most tracers need to be utilized early in the drug development process. Used at the right time, imaging biomarkers are able to inform the design of Phase II studies, including dose ranging and possibly patient selection, saving many months in development and millions of dollars in costs.”

    Answers from Big Data

  • “Clinical bioinformatics is the application of a data-driven, high-tech approach in clinical setting,” says Jerome Wojcik, Ph.D., CEO of Quartz Bio, a clinical bioinformatics service provider located in Plan-Les-Ouates, Switzerland. “We use clinical bioinformatics to adapt treatment to patients, that is, to identify cohorts that respond to the drug in a predictable manner,” says Dr. Wojcik.

    Pharmaceutical partners supply Quartz Bio with data collected in a course of clinical trials. The data (which may include information from protein and RNA expression, genotyping, molecular diagnostics, and flow cytometry studies) often exists in silos within a pharma company. To make sense of the data, Quartz Bio integrates heterogeneously formatted data, analyzes it for consistency, and identifies gaps and outliers.

    Dr. Wojcik’s team dedicates over 40% of the overall analysis time to the biomarker data management. This key step is crucial for the quality of the overall analysis. According to Quartz Bio, all the data-management processes are documented, auditable, and reproducible.

    Once the “Big Data” horde is adequately cleaned up, the team applies adaptive statistical methods to generate multiple hypotheses linking the drug action with subpopulations of patients. “Our challenge is to generate reliable hypotheses on a fairly small statistical patient sample, for example, a thousand patients, but using millions of biomarker datapoints,” continues Dr. Wojcik. “We do not rely on statistics alone. Graphical visualization adapted to the objectives of the study is necessary for interpretation of results.”

    In a recent project, Quartz Bio analyzed multiple oncology biomarkers, such as gene expression, circulating tumor cells, and immunohistochemistry, to identify patient cohorts that would most likely benefit from a novel treatment. Biomarker analysis revealed a subpopulation whose survival rate increased significantly over the population average, bringing a potential application of personalized medicine closer to reality.

     

 

Read Full Post »

%d bloggers like this: