Details

Value Creation in the Pharmaceutical Industry


Value Creation in the Pharmaceutical Industry

The Critical Path to Innovation
1. Aufl.

von: Alexander Schuhmacher, Markus Hinder, Oliver Gassmann

97,99 €

Verlag: Wiley-VCH
Format: PDF
Veröffentl.: 19.01.2016
ISBN/EAN: 9783527693436
Sprache: englisch
Anzahl Seiten: 508

DRM-geschütztes eBook, Sie benötigen z.B. Adobe Digital Editions und eine Adobe ID zum Lesen.

Beschreibungen

This practical guide for advanced students and decision-makers in the pharma and biotech industry presents key success factors in R&D along with value creators in pharmaceutical innovation. <br> A team of editors and authors with extensive experience in academia and industry and at some of the most prestigious business schools in Europe discusses in detail the innovation process in pharma as well as common and new research and innovation strategies. In doing so, they cover collaboration and partnerships, open innovation, biopharmaceuticals, translational medicine, good manufacturing practice, regulatory affairs, and portfolio management. Each chapter covers controversial aspects of recent developments in the pharmaceutical industry, with the aim of stimulating productive debates on the most effective and efficient innovation processes.<br> A must-have for young professionals and MBA students preparing to enter R&D in pharma or biotech as well as for students on a combined BA/biomedical and natural sciences program.
<p>List of Contributors XVII</p> <p>Foreword XXI</p> <p><b>1 Introduction to the Book 1</b><br /><i>Alexander Schuhmacher, Oliver Gassmann, and Markus Hinder</i></p> <p>Reference 8</p> <p><b>2 Global Epidemiological Developments 11</b><br /><i>Stephan Luther and Peter Schmitz</i></p> <p>2.1 Introduction 11</p> <p>2.2 Model of Epidemiological Transition 12</p> <p>2.3 Global Burden of Diseases 15</p> <p>2.3.1 Trends in the Distribution of Disease Burden 16</p> <p>2.4 Infectious Diseases 20</p> <p>2.4.1 (Re-)emerging Infectious Diseases 23</p> <p>2.4.2 Neglected Tropical Diseases 26</p> <p>2.5 Noncommunicable Diseases 29</p> <p>2.6 Antimicrobial Resistance 32</p> <p>2.7 Dynamics 35</p> <p>References 38</p> <p><b>3 The Value of Pharmaceutical Innovation: Concepts and Assessment 45</b><br /><i>Sam Salek and Paul Kamudoni</i></p> <p>3.1 Introduction 45</p> <p>3.2 Concepts and Definitions of Value 46</p> <p>3.3 Stakeholder’s Perspectives on Value 47</p> <p>3.3.1 Drug Regulatory Agencies 47</p> <p>3.3.2 Health Technology Assessment 47</p> <p>3.3.3 Patients 49</p> <p>3.3.4 Prescribers/Clinicians 49</p> <p>3.4 Recent Developments Influencing the Definition and Assessment of Value 50</p> <p>3.5 Recommendations: Implications for R&D 51</p> <p>3.6 Discussion 52</p> <p>3.7 Conclusion 56</p> <p>References 57</p> <p><b>4 A Review of the Pharmaceutical R&D Efficiency: Costs, Timelines, and Probabilities 61</b><br /><i>Alexander Schuhmacher, Oliver Gassmann, and Markus Hinder</i></p> <p>4.1 Introduction 61</p> <p>4.2 The Historical Perspective 62</p> <p>4.3 The R&D Phase Model 63</p> <p>4.4 The Low R&D Success Rates 63</p> <p>4.5 The Long R&D Time Intervals 67</p> <p>4.6 The High Cost of Pharmaceutical R&D 71</p> <p>4.7 The Reduced R&D Efficiency 73</p> <p>4.8 Can an Increase in R&D Value Compensate the Reduced R&D Efficiency? 76</p> <p>References 78</p> <p><b>5 Financing Pharmaceutical Innovation 81</b><br /><i>Sviataslau Sivagrakau</i></p> <p>5.1 Introduction 81</p> <p>5.2 Measuring Innovation: Categories of New Drugs 84</p> <p>5.3 Productivity of Pharmaceutical Industry throughout Time 86</p> <p>5.4 Measuring the Cost of Developing New Medicines 87</p> <p>5.5 Funding Drug Development: a Global Endeavor 91</p> <p>5.6 Public and Private Funds: Complementary Finance for Drug Development 95</p> <p>5.7 How Commercial Drug Development Projects Are Financed Today: Big Firms, Small Firms, andTheir Cooperation 97</p> <p>5.8 Public Health Economics and Financing Pharmaceutical Innovation 99</p> <p>5.9 Conclusion 101</p> <p>Acknowledgment 102</p> <p>References 102</p> <p><b>6 Challenges and Options for Drug Discovery 107</b><br /><i>Werner Kramer</i></p> <p>6.1 Introduction 107</p> <p>6.2 Paradigm Shifts of R&D Organizations 108</p> <p>6.3 Productivity of Drug Discovery 109</p> <p>6.4 IsThere an Innovation Gap in Biomedical Research? 111</p> <p>6.4.1 To Go for First in Class or Best in Class 112</p> <p>6.4.2 HowWe Define Medical Innovation? 112</p> <p>6.5 Why Did Drug Candidates Fail? 113</p> <p>6.5.1 Why Is the Dropout Rate So High in Early Clinical Development? 115</p> <p>6.5.1.1 Drug Behavior In Vivo: Role of Transport Proteins 115</p> <p>6.5.1.2 Hypes and Lack of Scientific Thoroughness 116</p> <p>6.6 Implications from the "Lessons Learnt" for Future Drug Discovery Research 123</p> <p>6.6.1 Organization of Drug Discovery and Development 123</p> <p>6.6.2 Elucidation of the Physiological Validity of a Target for the Human Disease 125</p> <p>6.6.2.1 Extensive Inquiry of (All) Published Data of a Target or Pathway 125</p> <p>6.6.2.2 Integrative Knowledge Management 127</p> <p>6.6.2.3 Demonstration of the Involvement of a Target in Human Disease 128</p> <p>6.6.2.4 A Stringent and Comprehensive Test Sequence 132</p> <p>6.6.2.5 Translational Clinical Trials 135</p> <p>Acknowledgment 136</p> <p>References 136</p> <p><b>7 Translational Medicine: Enabling the Proof of Concepts 141</b><br /><i>Gezim Lahu and John Darbyshire</i></p> <p>7.1 Introduction 141</p> <p>7.2 Translational Medicine and Its Role/Value in Early Development 143</p> <p>7.3 Knowledge Generation 144</p> <p>7.4 Types of Data, Experiments, and Tools Needed to Move from Basic Research to Early Clinical Development 144</p> <p>7.4.1 Dose Selection 145</p> <p>7.4.2 Animal Models 146</p> <p>7.4.3 Fraction of NOAEL and Efficacious Dose 149</p> <p>7.4.4 Allometric Scaling and PBPK 150</p> <p>7.4.5 Physiologically Based Pharmacokinetic Models PBPK 151</p> <p>7.4.6 Pharmacokinetic and Pharmacodynamic Modeling 151</p> <p>7.5 FIM (Dose Escalation and MTD) 153</p> <p>7.6 Proof of Concept (PoC) 154</p> <p>Summary 156</p> <p>References 157</p> <p><b>8 Preclinical Safety and Risk Assessment 161</b><br /><i>Paul Germann and Rob Caldwell</i></p> <p>8.1 Introduction 161</p> <p>8.2 Test Systems 161</p> <p>8.2.1 In Silico Analysis 161</p> <p>8.2.2 In Vitro Experiments 162</p> <p>8.3 Case Study: hERG Assay 163</p> <p>8.3.1 In Vivo Experiments 164</p> <p>8.4 The Preclinical "Package" during the Development of an NME 165</p> <p>8.5 Factors Influencing the Preclinical Data Set 166</p> <p>8.5.1 Timing and Costs 167</p> <p>8.5.2 Intended Clinical Application Route 167</p> <p>8.5.3 Treatment Duration and Treatment Frequency 167</p> <p>8.5.4 Clinical Indication 167</p> <p>8.5.5 Ongoing Changes of the Regulatory Landscape 168</p> <p>8.5.6 New Drug Formats 168</p> <p>8.6 Translation into Humans:The "TherapeuticWindow" 169</p> <p>8.7 Influence of Intended Therapeutic Use on the Risk Assessment (RA) 169</p> <p>8.8 Deep Dive Case Study: Safety Assessment of Biological Drug Formats 170</p> <p>8.9 NBE Case Study 1 175</p> <p>8.10 NBE Case Study 2 175</p> <p>8.11 Carcinogenicity Risk Assessment for Marketed Drugs 176</p> <p>8.12 Treatment Duration 178</p> <p>8.13 Conclusion – the "Art" of Preclinical Safety: Summarizing the Concept of Hazard Identification and Description, Risk Assessment, and Risk Management 179</p> <p>Acknowledgment 179</p> <p>Disclosures 180</p> <p>References 180</p> <p><b>9 Developing Commercial Solutions for Therapeutic Proteins 183</b><br /><i>Galina Hesse</i></p> <p>9.1 Introduction 183</p> <p>9.2 Developing Commercial Solutions for Therapeutic Proteins 184</p> <p>9.2.1 Defining a Target Product Profile 184</p> <p>9.2.2 Developing Formulations for Therapeutic Proteins 186</p> <p>9.2.3 Testing Formulations for Therapeutic Proteins 188</p> <p>9.2.4 Development of Primary Containers 188</p> <p>9.2.5 Development of Application Systems 190</p> <p>9.3 Quality by Design 192</p> <p>9.4 Examples for Innovations in Manufacture of Sterile Pharmaceutical Products 194</p> <p>9.5 Summary 197</p> <p>List of FDA/ICH Guidances Referenced 198</p> <p>Disclaimer 199</p> <p>References 199</p> <p><b>10 The Evolution of Clinical Development: From Technical Success to Clinical Value Creation 203</b><br /><i>Markus Hinder and Alexander Schuhmacher</i></p> <p>10.1 Introduction 203</p> <p>10.2 CD: Changes and Challenges 204</p> <p>10.2.1 Clinical Endpoints: From Symptom-Oriented Endpoints to Hard and Predefined Endpoints 204</p> <p>10.2.2 Determination and Quantification of Risks 205</p> <p>10.2.3 Assessment of Medical Progress in Context of Available Therapeutic Options 206</p> <p>10.2.3.1 EbM 206</p> <p>10.2.3.2 Health Economics, Pharmacoeconomics, and the Fourth Hurdle 207</p> <p>10.2.3.3 Results of These Changes and Challenges 208</p> <p>10.3 Technical Success and Clinical Value Creation in CD in the Future 208</p> <p>10.3.1 Established and Novel Approaches to Determine the Dose–Exposure–Response Relationship 210</p> <p>10.3.2 Comparators 212</p> <p>10.3.3 Patient Stratification to Increase Treatment Response and Benefit and Reduce Risk 212</p> <p>10.3.4 New Operational Tools to Succeed in Trials with Increased Complexity, Special Populations, or Large Size 213</p> <p>10.3.5 Collaboration and Outsourcing as Tools toWork in Networks 214</p> <p>10.3.6 Collaboration across Sectors and Industries to Boost the NextWave of Innovation 215</p> <p>Disclaimer 218</p> <p>References 218</p> <p><b>11 Translational Development 225</b><br /><i>Nigel McCracken</i></p> <p>11.1 Introduction 225</p> <p>11.1.1 Legacy 226</p> <p>11.2 Translational Development 227</p> <p>11.2.1 TP 228</p> <p>11.2.2 Translational Toolkit 229</p> <p>11.3 Dose Optimization 230</p> <p>11.3.1 Physicochemical Properties 231</p> <p>11.3.2 Target Affinity and Selectivity 231</p> <p>11.3.3 Clearance 231</p> <p>11.3.4 Prediction of Human Dose 232</p> <p>11.4 Pharmacogenomics 233</p> <p>11.4.1 Patient Segmentation 233</p> <p>11.4.2 Disease Segmentation 234</p> <p>11.4.3 Utility 237</p> <p>11.5 Biomarker Development 238</p> <p>11.5.1 Biomarker Activities 239</p> <p>11.5.2 Assessing the Opportunity 239</p> <p>11.6 Systems Pharmacology 240</p> <p>11.7 Rational Drug Development 241</p> <p>11.8 Concluding Remarks 242</p> <p>References 242</p> <p><b>12 Forty Years of Innovation in Biopharmaceuticals – Will the Next 40 Years Be as Revolutionary? 245</b><br /><i>Mathias Schmidt, Sanjay Patel, Petter Veiby, Qiang Liu, and Michael Buckley</i></p> <p>12.1 Introduction 245</p> <p>12.1.1 The Value Proposition of Biologics 246</p> <p>12.1.1.1 The Patient Perspective 246</p> <p>12.1.1.2 The Pharmaceutical Industry’s Perspective 248</p> <p>12.1.2 Biosimilars: A Blessing or aThreat to Innovation? 250</p> <p>12.1.3 Further Innovation in Biologics – Incremental or Revolutionary? 252</p> <p>12.2 The Evolution of Biologics Manufacturing 252</p> <p>12.2.1 Introduction 252</p> <p>12.2.2 CHO Cells: The Industry Workhorse 253</p> <p>12.2.3 Protein Production Strategies 253</p> <p>12.2.4 The Impact of Increasing Titers on Manufacturing Facilities 255</p> <p>12.2.5 Protein Purification Platforms 256</p> <p>12.2.6 Conclusion: WhatWill the Next 40 Years of Innovation Bring? 258</p> <p>12.3 The Evolution of Alternative Scaffolds 259</p> <p>12.3.1 Novel Small Protein Scaffolds 260</p> <p>12.3.2 Single-Chain Fragment Variables and Diabodies 260</p> <p>12.3.3 Single-Domain Antibodies 261</p> <p>12.3.4 Nonantibody Scaffolds 261</p> <p>12.3.5 Bispecific Single-Chain Fragment Variables and Diabodies 263</p> <p>12.3.6 Other Bispecific Antibody Formats 264</p> <p>12.4 Antibody-Drug Conjugates 265</p> <p>12.5 The Next Wave of Biologics 270</p> <p>12.5.1 Orally Available Biologics 271</p> <p>12.5.2 Biologics That Enter the Cytoplasm 271</p> <p>12.5.3 Biologics That Pass the Blood–Brain Barrier 272</p> <p>12.5.4 Translational Medicine as Driver of Innovation 272</p> <p>Disclaimer 273</p> <p>References 273</p> <p><b>13 Vaccines: Where Inertia, Innovation, and Revolution Create Value, Simultaneously and Quietly 277</b><br /><i>Pierre A. Morgon and Hannah Nawi</i></p> <p>13.1 Introduction 277</p> <p>13.2 TheWorld of Vaccines 278</p> <p>13.2.1 What Are Vaccines? 278</p> <p>13.2.2 Current Vaccines Are Mainly Prophylactic: Curative Vaccines Are Emerging 278</p> <p>13.2.3 Drivers to Immunize: Individual and Collective 280</p> <p>13.2.4 The Pivotal Role of Recommendations 280</p> <p>13.3 The Vaccine Market: Substantial, Fast Growing, with Intense and Concentrated Competition 281</p> <p>13.4 The Vaccine Industry: Domination of the Heavyweights, for Now… 282</p> <p>13.4.1 Barriers to Entry: From R&D Risk to Capital Intensiveness 290</p> <p>13.4.2 Five Forces Analysis: Competitive Intensiveness and Downstream Hurdles 291</p> <p>13.4.2.1 Acceptability 291</p> <p>13.4.2.2 Accessibility 292</p> <p>13.4.2.3 Availability 293</p> <p>13.4.2.4 Affordability 293</p> <p>13.5 New Vaccine Developments: Strategic Trends and Why Innovation Is Needed All along the Value Chain 295</p> <p>13.5.1 Where Is Innovation Needed? R&D 296</p> <p>13.5.2 Where Is Innovation Needed? Manufacturing and Product Improvement 301</p> <p>13.5.3 Where Is Innovation Needed? Acceptability 301</p> <p>13.5.4 Where Is Innovation Needed? Accessibility, Both as a Function of Supply (Availability) and Logistics 302</p> <p>13.5.5 Affordability and Sustainability 303</p> <p>13.6 WhereWill Innovation Come from? Strategy and Players 304</p> <p>13.6.1 Take-Home Messages 305</p> <p>References 306</p> <p><b>14 The Patient-Centric Pharma Company: Evolution, Reboot, or Revolution? 309</b><br /><i>Pierre A. Morgon</i></p> <p>14.1 Introduction 309</p> <p>14.2 Health, Always… 310</p> <p>14.3 The Mission of the Healthcare Industry 310</p> <p>14.4 Megatrends Affecting the Strategic Scorecard of the Healthcare Industry 312</p> <p>14.5 Focus on the Societal Trends and Their Consequences for the Management of Healthcare Innovation 314</p> <p>14.6 The DNA of the Healthcare Industry: R&D and the Management of Innovation 316</p> <p>14.7 Societal Expectations for Personalized Medicine 318</p> <p>14.8 New Players Contributing to Information Management to Substantiate Value Propositions for NovelTherapies 319</p> <p>14.9 The Role of the Key Stakeholders in Shaping a New Regulatory Framework 323</p> <p>14.10 The Consequences for the Healthcare Industry in Terms of Governance and Capabilities 325</p> <p>14.11 The Sustainable Path Forward for the Healthcare Industry 329</p> <p>14.11.1 Take-Home Messages 331</p> <p>References 332</p> <p><b>15 The Pharmaceutical Industry is Opening Its R&D Boundaries 335</b><br /><i>Alexander Schuhmacher and Ulrich A. K. Betz</i></p> <p>15.1 Introduction 335</p> <p>15.2 Open Innovation versus Closed Innovation 336</p> <p>15.3 Business Models in an Open Innovation Framework 341</p> <p>15.4 Open Innovation Processes 342</p> <p>15.5 Capabilities and Attitudes Enabling Open Innovation 344</p> <p>15.6 Open Innovation in the Pharmaceutical Industry 345</p> <p>15.6.1 The More Traditional Elements of Open Innovation 345</p> <p>15.6.1.1 Target Scouting 345</p> <p>15.6.1.2 Research Collaborations 346</p> <p>15.6.1.3 Drug Licensing 346</p> <p>15.6.1.4 Outsourcing 348</p> <p>15.6.1.5 Joint Ventures 349</p> <p>15.6.2 The Newer Concepts of Open Innovation 349</p> <p>15.6.2.1 New Frontier Science 350</p> <p>15.6.2.2 Drug Discovery Alliances 350</p> <p>15.6.2.3 Private–Public Partnerships 351</p> <p>15.6.2.4 Innovation Incubator 351</p> <p>15.6.2.5 Virtual R&D 352</p> <p>15.6.2.6 Crowdsourcing 353</p> <p>15.6.2.7 Open Source Innovation 355</p> <p>15.6.2.8 Innovation Camps 355</p> <p>15.6.2.9 Fluctuating Open Teams 356</p> <p>15.7 New Business Models in View of the Potential of Open Innovation 356</p> <p>15.7.1 General Trends in the Pharmaceutical Industry 356</p> <p>15.8 Outlook 358</p> <p>References 359</p> <p><b>16 Out-Licensing in Pharmaceutical Research and Development 363</b><br /><i>Oliver Gassmann, Carol A. Krech, Martin A. Bader, and Gerrit Reepmeyer</i></p> <p>16.1 Introduction 363</p> <p>16.2 Performance-Based R&D Collaborations on the Rise 364</p> <p>16.3 The Impact of Collaborations on the Value Chain 365</p> <p>16.4 Generating Value from Pipeline Assets by Out-Licensing 367</p> <p>16.5 Pharmaceutical Companies’ Resistance toward Out-Licensing 372</p> <p>16.6 Managing Out-Licensing at Novartis: A Case Study 372</p> <p>16.6.1 Out-Licensing as a 10-Step Process 373</p> <p>16.6.2 Out-Licensing Contract Design 375</p> <p>16.6.3 Structure of the Out-Licensing Collaboration with Speedel 375</p> <p>16.7 Future Directions and Trends 377</p> <p>References 378</p> <p><b>17 Trends and Innovations in Pharmaceutical R&D Outsourcing 383</b><br /><i>Antal K. Hajos</i></p> <p>17.1 Introduction 383</p> <p>17.2 Drivers to the Use of Outsourcing 383</p> <p>17.2.1 Overview on the CRO Market 383</p> <p>17.2.2 Core versus Noncore Activities 387</p> <p>17.3 Genesis of Outsourcing in the Twentieth Century: From Commodity to Contribution 388</p> <p>17.3.1 Outsourcing Portfolio and the Move to Full-Service Provision 388</p> <p>17.3.2 Globalization and the Emerging Market Hype 389</p> <p>17.3.3 Procurement Takes over the Outsourcing Function 391</p> <p>17.4 Current and Future Trends in Outsourcing: From Contribution to Innovation 392</p> <p>17.4.1 How Has Outsourcing Itself Innovated and What Are the Future Trends? 392</p> <p>17.4.2 How Does andWill Outsourcing Contribute to Innovation? 394</p> <p>17.5 Discussion and Conclusion 395</p> <p>References 398</p> <p><b>18 New Innovation Models in Pharmaceutical R&D 401</b><br /><i>Alexander Schuhmacher, Oliver Gassmann, and Markus Hinder</i></p> <p>18.1 Introduction 401</p> <p>18.2 Some AttemptsThatWere Recommended in the Past 402</p> <p>18.3 The Increasing Pipeline Size 403</p> <p>18.4 The Reduction of R&D Investments 404</p> <p>18.5 The Opening of the R&D Processes 407</p> <p>18.6 The Challenge with the Return on Investment 411</p> <p>18.7 Changing the R&D Processes Is Not Enough 412</p> <p>18.8 What Is the Best R&D Model? 413</p> <p>References 414</p> <p><b>19 The Influence of Leadership Paradigms and Styles on Pharmaceutical Innovation 416</b><br /><i>Aubyn Howard</i></p> <p>19.1 Introduction 417</p> <p>19.2 What Is Your Concept or Model of Good Leadership? 419</p> <p>19.3 Approaches to Leadership Modeling and Profiling 420</p> <p>19.3.1 Personality Types 421</p> <p>19.3.2 Behavioral Preferences 421</p> <p>19.3.3 Developmental Stages 421</p> <p>19.3.4 Competency Frameworks 421</p> <p>19.4 The Developmental Approach to Leadership Paradigms and Styles 422</p> <p>19.5 Inner and Outer Leadership 424</p> <p>19.6 Dynamics of How Leadership Paradigms Evolve 425</p> <p>19.6.1 Magic–Animistic 426</p> <p>19.6.2 Impulsive–Egocentric 427</p> <p>19.6.3 Conformist–Absolutist 428</p> <p>19.6.4 Achievement–Multiplistic 429</p> <p>19.6.5 Pluralistic–Relativistic 430</p> <p>19.6.6 Evolutionary–Systemic 432</p> <p>19.7 Leadership at Different Levels within Pharma 433</p> <p>19.8 Optimizing Innovation in Different Organizational Models and Cultures 437</p> <p>19.9 How DoWe Support the Development of Evolutionary Leaders? 439</p> <p>19.10 What Does It Mean to Operate from the Evolutionary Paradigm? 440</p> <p>19.11 Leadership and Personal Mastery 441</p> <p>19.12 Building an Evolutionary Bridge to Release Innovation 442</p> <p>19.13 Conclusions 445</p> <p>References 446</p> <p><b>20 The Role of Modern Portfolio Management in Pharma Innovation 449</b><br /><i>Joachim M. Greuel and Axel Wiest</i></p> <p>20.1 Introduction 449</p> <p>20.2 Challenges in R&D and the Origin of Pharmaceutical Portfolio Management 450</p> <p>20.3 Goals and Metrics of Portfolio Management 451</p> <p>20.4 Portfolio Management as Enabler of Innovation 456</p> <p>20.5 Modern Portfolio Management Integrates In-House R&D, Business Development, and M&A 457</p> <p>References 458</p> <p><b>21 Patent Management Throughout the Innovation Life Cycle 461</b><br /><i>Martin A. Bader and Oliver Gassmann</i></p> <p>21.1 Introduction 461</p> <p>21.2 The Changing Role of Patents: From Legal to Strategic 462</p> <p>21.3 The Patent Life Cycle Management Model 467</p> <p>21.3.1 Exploration 468</p> <p>21.3.2 Generation 469</p> <p>21.3.3 Protection 469</p> <p>21.3.4 Optimization 470</p> <p>21.3.5 Decline 470</p> <p>21.4 Example: Managing IP Rights at Bayer 471</p> <p>21.5 Concluding Remarks 472</p> <p>References 473</p> <p>Index 475</p>
<p><b>Prof. Dr. Alexander Schuhmacher</b> is a professor for R&D management, Vice Dean of the Faculty of Applied Chemistry and Senator at Reutlingen University. And he is Director for R&D performance metrics and business model innovation at Bioscience Valuation. Before joining the academic world, he worked 14 years in the pharmaceutical industry in various functions in R&D, such as in R&D portfolio management and strategic planning. He studied biology at the University of Constance (Germany), Pharmaceutical Medicine at Witten-Herdecke University (Germany) and he is also a graduate of the Executive MBA program at the University of St. Gallen (Switzerland).</p> <p><b>Prof. Dr. Markus Hinder</b> studied medicine at the Universities of Heidelberg, Paris and Zurich and obtained a doctoral degree in pharmacology from Heidelberg University. After graduation he trained in clinical pharmacology, cardiology and emergency medicine. Before joining Novartis he held leadership positions in clinical pharmacology, translational medicine, clinical development, medical affairs and project management. Markus is a professor at Cardiff University/ Hochschule Fresenius, reviewer for several journals and associate editor for the Journal of Translational Medicine.</p> <p><b>Prof. Dr. Oliver Gassmann</b> is a Professor for technology and innovation management at St. Gallen University, where he chairs the Institute of Technology Management. His teaching activities include several executive MBA programs. He has written or edited 18 books and published more than 300 journal articles on technology and innovation management. Until 2002 he headed the R&D department of Schindler. The main focus of his research lies in open innovation and global innovation processes. He is the 1998 recipient of the RADMA Prize and in 2009 was elected among the top 50 researchers by IAMOT, the International Association for Management of Technology.</p>

Diese Produkte könnten Sie auch interessieren:

Hot-Melt Extrusion
Hot-Melt Extrusion
von: Dennis Douroumis
PDF ebook
136,99 €
Hot-Melt Extrusion
Hot-Melt Extrusion
von: Dennis Douroumis
EPUB ebook
136,99 €
Kunststoffe
Kunststoffe
von: Wilhelm Keim
PDF ebook
99,99 €