IELTS Reading: Nghiên Cứu Di Truyền Học Chữa Bệnh – Đề Thi Mẫu Có Đáp Án Chi Tiết

Mở Bài

Chủ đề nghiên cứu di truyền học và khả năng chữa trị bệnh tật đang trở thành một trong những đề tài phổ biến nhất trong kỳ thi IELTS Reading hiện nay. Với sự phát triển vượt bậc của khoa học y sinh, đề tài này xuất hiện thường xuyên trong các đề thi IELTS chính thức, đặc biệt ở Passage 2 và Passage 3 với độ khó từ trung bình đến cao.

Bài viết này cung cấp cho bạn một bộ đề thi IELTS Reading hoàn chỉnh với 3 passages được thiết kế tăng dần độ khó, từ Easy (Band 5.0-6.5) đến Medium (Band 6.0-7.5) và Hard (Band 7.0-9.0). Bạn sẽ được thực hành với 40 câu hỏi đa dạng các dạng bài giống như trong kỳ thi thật, kèm theo đáp án chi tiết và giải thích cụ thể.

Đặc biệt, bài viết còn cung cấp phân tích từ vựng chuyên ngành, chiến lược làm bài hiệu quả và những kỹ thuật paraphrase quan trọng giúp bạn tự tin đạt band điểm cao. Đây là tài liệu phù hợp cho tất cả học viên từ band 5.0 trở lên, đặc biệt hữu ích cho những ai đang hướng tới band 7.0-8.0.

Hướng Dẫn Làm Bài IELTS Reading

Tổng Quan Về IELTS Reading Test

IELTS Reading Test bao gồm 3 passages với tổng cộng 40 câu hỏi phải hoàn thành trong 60 phút. Đây là bài kiểm tra khả năng đọc hiểu, phân tích thông tin và quản lý thời gian của thí sinh.

Phân bổ thời gian khuyến nghị:

  • Passage 1: 15-17 phút (độ khó thấp nhất)
  • Passage 2: 18-20 phút (độ khó trung bình)
  • Passage 3: 23-25 phút (độ khó cao nhất)

Lưu ý dành 2-3 phút cuối để chuyển đáp án lên phiếu trả lời. Không có thời gian thêm cho việc này trong phòng thi.

Các Dạng Câu Hỏi Trong Đề Này

Đề thi mẫu này bao gồm 7 dạng câu hỏi phổ biến nhất:

  1. Multiple Choice – Trắc nghiệm nhiều lựa chọn
  2. True/False/Not Given – Xác định thông tin đúng/sai/không đề cập
  3. Yes/No/Not Given – Xác định quan điểm tác giả
  4. Matching Headings – Nối tiêu đề với đoạn văn
  5. Sentence Completion – Hoàn thành câu
  6. Summary Completion – Hoàn thành tóm tắt
  7. Matching Features – Nối thông tin với đặc điểm

IELTS Reading Practice Test

PASSAGE 1 – The Dawn of Genetic Medicine

Độ khó: Easy (Band 5.0-6.5)

Thời gian đề xuất: 15-17 phút

The story of genetic research transforming medical treatment began in the mid-20th century when scientists first discovered the structure of DNA. This groundbreaking discovery by James Watson and Francis Crick in 1953 laid the foundation for understanding how genetic information is stored and transmitted in living organisms. Today, more than seventy years later, we are witnessing a revolutionary era in medicine where scientists can not only read the genetic code but also edit it to cure diseases that were once considered incurable.

Genetic disorders affect millions of people worldwide. These conditions result from abnormalities in an individual’s DNA, which can be inherited from parents or occur spontaneously during a person’s lifetime. Some well-known genetic disorders include sickle cell disease, cystic fibrosis, and Huntington’s disease. For decades, doctors could only manage the symptoms of these conditions, but they could not address the root cause – the faulty genes themselves. This situation has changed dramatically with the advent of gene therapy and gene editing technologies.

One of the most significant breakthroughs in genetic medicine is gene therapy, a technique that involves introducing healthy genes into a patient’s cells to replace or compensate for defective genes. The first successful gene therapy trial took place in 1990 when a four-year-old girl with severe combined immunodeficiency (SCID) received treatment. Scientists extracted white blood cells from her body, inserted a functional gene to correct the deficiency, and returned the modified cells to her body. Although the initial results were modest, this pioneering treatment demonstrated that genetic intervention was possible.

The development of CRISPR-Cas9 technology in 2012 marked another milestone in genetic research. This gene-editing tool works like molecular scissors, allowing scientists to cut DNA at specific locations and either remove harmful genetic sequences or insert new ones. The precision and relative simplicity of CRISPR have made it accessible to researchers worldwide. Unlike earlier gene-editing methods that were expensive, time-consuming, and often imprecise, CRISPR can target specific genes with remarkable accuracy. This technology has opened up possibilities that seemed like science fiction just a decade ago.

In recent years, several genetic treatments have moved from laboratory experiments to clinical trials and even commercial availability. In 2017, the US Food and Drug Administration approved the first gene therapy for an inherited disease – a treatment for a rare form of inherited blindness. Patients who received this therapy showed significant improvement in their vision, with some able to see well enough to navigate independently for the first time in their lives. This success has encouraged pharmaceutical companies to invest heavily in developing genetic treatments for other conditions.

Blood disorders have been particularly amenable to genetic treatment. Sickle cell disease and beta-thalassemia, both caused by mutations in the hemoglobin gene, affect hundreds of thousands of people globally. Traditional treatment requires regular blood transfusions and medications, significantly impacting patients’ quality of life. However, new gene therapies have shown remarkable results. In one clinical trial, patients with severe sickle cell disease who received gene therapy were able to produce healthy red blood cells and experienced dramatic reductions in pain episodes and hospital visits.

Despite these encouraging developments, genetic medicine still faces significant challenges. The high cost of developing and administering gene therapies remains a major barrier. Current treatments can cost hundreds of thousands or even millions of dollars per patient, making them inaccessible to most people who need them. Additionally, long-term safety concerns persist because scientists are still learning about the potential side effects of altering human genes. There have been cases where gene therapy has triggered unexpected immune responses or caused genes to activate in unintended ways.

Ethical considerations also surround genetic research, particularly regarding germline editing – changes to genes that can be passed to future generations. While such editing could potentially eliminate genetic diseases from family lines, it raises profound questions about human enhancement and the possibility of creating genetic inequality. Many countries have imposed strict regulations or outright bans on germline editing in humans, though research continues in animals and human embryos not intended for pregnancy.

Looking ahead, researchers are optimistic that genetic medicine will become more refined, affordable, and widely available. Advances in artificial intelligence are helping scientists identify disease-causing genes more quickly and predict which genetic modifications will be most effective. Manufacturing processes are becoming more efficient, which should help reduce costs over time. As our understanding of the human genome deepens, we may discover ways to treat common diseases like diabetes, heart disease, and even certain forms of cancer through genetic interventions.

The transformation of genetic research into practical medical treatments represents one of humanity’s greatest scientific achievements. What began as a quest to understand the basic building blocks of life has evolved into a powerful toolkit for combating disease. While challenges remain, the progress made in recent decades suggests that genetic medicine will play an increasingly important role in healthcare, offering hope to patients with conditions that currently have no cure.

Nhà nghiên cứu đang tiến hành thí nghiệm chỉnh sửa gen CRISPR trong phòng thí nghiệm hiện đại với thiết bị công nghệ caoNhà nghiên cứu đang tiến hành thí nghiệm chỉnh sửa gen CRISPR trong phòng thí nghiệm hiện đại với thiết bị công nghệ cao

Questions 1-13

Questions 1-5: Multiple Choice

Choose the correct letter, A, B, C, or D.

1. The discovery of DNA structure in 1953 was important because it:

  • A. cured genetic diseases immediately
  • B. helped scientists understand genetic information storage
  • C. eliminated the need for medical research
  • D. proved all diseases were genetic

2. According to the passage, genetic disorders:

  • A. only affect children
  • B. can only be inherited from parents
  • C. may be inherited or occur spontaneously
  • D. are always fatal

3. The first successful gene therapy trial in 1990:

  • A. completely cured the patient
  • B. showed that genetic intervention was possible
  • C. failed to produce any results
  • D. was abandoned due to safety concerns

4. CRISPR-Cas9 technology is compared to:

  • A. molecular scissors
  • B. a microscope
  • C. a blood test
  • D. an X-ray machine

5. The passage suggests that gene therapy for inherited blindness:

  • A. had no effect on patients
  • B. made some patients see better
  • C. caused additional health problems
  • D. was rejected by authorities

Questions 6-9: True/False/Not Given

Do the following statements agree with the information given in the passage?

Write:

  • TRUE if the statement agrees with the information
  • FALSE if the statement contradicts the information
  • NOT GIVEN if there is no information on this

6. Watson and Crick discovered DNA in the 21st century.

7. Sickle cell disease can be inherited from parents.

8. All gene therapy treatments are affordable for most patients.

9. Gene therapy clinical trials have shown positive results for blood disorders.

Questions 10-13: Sentence Completion

Complete the sentences below.

Choose NO MORE THAN TWO WORDS from the passage for each answer.

10. Gene therapy involves introducing healthy genes to replace or compensate for __ genes.

11. CRISPR technology is more accessible than earlier methods because of its precision and relative __.

12. Many countries have imposed strict regulations on __ editing in humans.

13. Scientists are using __ __ to identify disease-causing genes more quickly.


PASSAGE 2 – Molecular Mechanisms and Therapeutic Applications

Độ khó: Medium (Band 6.0-7.5)

Thời gian đề xuất: 18-20 phút

The molecular basis of genetic diseases involves complex interactions between genes, proteins, and cellular processes. Understanding these mechanisms has been crucial for developing targeted therapies that can address the underlying causes of disease rather than merely alleviating symptoms. Recent advances in molecular biology and biotechnology have provided researchers with unprecedented tools to manipulate genetic material with precision, opening new therapeutic avenues that were unimaginable just two decades ago.

Gene expression, the process by which information from a gene is used to synthesize functional gene products such as proteins, represents a critical control point for therapeutic intervention. Many diseases result not from the absence of a gene but from its inappropriate expression – either too much, too little, or at the wrong time or place in the body. Scientists have developed several strategies to modulate gene expression. Antisense oligonucleotides, short DNA or RNA molecules that bind to messenger RNA (mRNA), can prevent the production of disease-causing proteins. This approach has proven successful in treating spinal muscular atrophy, a severe neuromuscular disorder that is the leading genetic cause of infant mortality.

The human immune system presents both opportunities and challenges for genetic medicine. On one hand, immune cells are relatively accessible and can be modified outside the body before being returned to the patient – an approach called ex vivo gene therapy. Chimeric antigen receptor (CAR) T-cell therapy exemplifies this strategy. In this treatment, scientists extract a patient’s T-cells, engineer them to express specialized receptors that recognize cancer cells, and infuse the modified cells back into the patient. These engineered immune cells can then identify and destroy cancer cells throughout the body. CAR T-cell therapies have achieved remarkable remission rates in certain blood cancers, though their effectiveness against solid tumors remains limited.

However, the immune system can also impede genetic treatments. When viral vectors – modified viruses used to deliver therapeutic genes – are introduced into the body, they may trigger immune responses that neutralize the treatment or cause adverse reactions. Some patients have pre-existing antibodies against commonly used viral vectors, making them ineligible for certain gene therapies. Researchers are addressing this challenge by developing novel delivery systems, including non-viral vectors and engineered viruses that evade immune detection. Lipid nanoparticles, which encapsulate genetic material in protective fatty shells, have shown promise as alternative delivery vehicles, particularly for RNA-based therapies.

Genetic heterogeneity – the phenomenon where different genetic mutations produce similar disease phenotypes – complicates the development of universal treatments. For instance, more than 2,000 different mutations in the CFTR gene can cause cystic fibrosis, and each mutation may require a slightly different therapeutic approach. This has led to the concept of precision medicine, where treatments are tailored to a patient’s specific genetic profile. Pharmacogenomics, the study of how genes affect drug responses, helps predict which patients will benefit from particular medications and which may experience adverse effects. This individualized approach maximizes therapeutic efficacy while minimizing risks and unnecessary treatments.

In vivo gene therapy, where genetic modifications occur directly within the patient’s body, offers advantages for treating diseases affecting inaccessible tissues like the brain or liver. However, achieving efficient gene delivery to specific organs while avoiding off-target effects in other tissues remains technically challenging. Recent innovations in vector design have improved tissue specificity. For example, adeno-associated viruses (AAV) can be engineered with different capsid proteins that determine which cell types they can infect. By selecting appropriate AAV serotypes, researchers can preferentially deliver genes to hepatocytes, neurons, or muscle cells, depending on the therapeutic objective.

Base editing and prime editing represent the latest evolution of gene-editing technology, offering even greater precision than standard CRISPR systems. While conventional CRISPR creates double-strand breaks in DNA, which cells repair through processes that can introduce errors, base editors chemically modify individual DNA letters without cutting both strands. Prime editors can make even more complex changes, including insertions, deletions, and replacing multiple letters simultaneously. These refined tools reduce unintended mutations and expand the range of genetic alterations possible, potentially enabling the correction of a broader spectrum of disease-causing mutations.

The regulatory landscape for genetic therapies continues to evolve as these treatments move from experimental protocols to standard clinical practice. Regulatory agencies must balance the urgent need for life-saving treatments against the necessity of ensuring long-term safety. The approval process for gene therapies typically requires extensive preclinical studies, multiple phases of clinical trials, and post-marketing surveillance to detect late-emerging side effects. Some regulatory authorities have created expedited pathways for promising therapies addressing unmet medical needs, allowing patients to access treatments more quickly while continuing to monitor safety and efficacy.

Manufacturing scalability poses another significant hurdle for widespread adoption of genetic medicines. Producing viral vectors or engineering immune cells requires specialized facilities and highly skilled personnel, limiting the number of patients who can be treated simultaneously. Each batch must undergo rigorous quality control to ensure consistency, purity, and potency. As demand for genetic therapies increases, biopharmaceutical companies are investing in automated production systems and larger manufacturing capacity. Some companies are developing off-the-shelf therapies using donor cells rather than a patient’s own cells, which could dramatically reduce production costs and treatment timelines.

Looking toward the future, convergence of genetic medicine with other cutting-edge technologies promises to accelerate progress. Artificial intelligence algorithms can analyze vast genomic datasets to identify disease-associated genes and predict optimal editing strategies. Organ-on-chip systems provide more realistic preclinical testing platforms than traditional cell cultures or animal models, potentially improving the success rate of clinical translation. Nanotechnology offers new possibilities for precise drug delivery and real-time monitoring of therapeutic outcomes. As these technologies mature and integrate, the distinction between genetic, pharmaceutical, and regenerative medicine may blur, creating hybrid approaches that leverage the strengths of each modality.

Sơ đồ minh họa chi tiết cơ chế hoạt động của công nghệ chỉnh sửa gen CRISPR-Cas9 trong điều trị bệnh di truyềnSơ đồ minh họa chi tiết cơ chế hoạt động của công nghệ chỉnh sửa gen CRISPR-Cas9 trong điều trị bệnh di truyền

Questions 14-26

Questions 14-18: Yes/No/Not Given

Do the following statements agree with the views of the writer in the passage?

Write:

  • YES if the statement agrees with the views of the writer
  • NO if the statement contradicts the views of the writer
  • NOT GIVEN if it is impossible to say what the writer thinks about this

14. Understanding molecular mechanisms is essential for developing treatments that address disease causes.

15. All genetic diseases result from the complete absence of a gene.

16. CAR T-cell therapy is equally effective against all types of cancer.

17. Pre-existing antibodies can prevent some patients from receiving certain gene therapies.

18. Animal models are superior to organ-on-chip systems for testing.

Questions 19-23: Matching Headings

The passage has ten paragraphs (1-10). Choose the correct heading for paragraphs 2, 4, 5, 7, and 9 from the list of headings below.

List of Headings:

  • i. The challenge of manufacturing genetic therapies
  • ii. Modulating gene expression for treatment
  • iii. Future integration of multiple technologies
  • iv. Immune system obstacles to genetic medicine
  • v. Personalized medicine and genetic variation
  • vi. Advanced gene-editing techniques
  • vii. The role of viral vectors
  • viii. Regulatory approval processes
  • ix. Cost concerns in genetic research
  • x. Direct gene therapy within the body

19. Paragraph 2 ____

20. Paragraph 4 ____

21. Paragraph 5 ____

22. Paragraph 7 ____

23. Paragraph 9 ____

Questions 24-26: Summary Completion

Complete the summary below.

Choose NO MORE THAN TWO WORDS from the passage for each answer.

Base editing and prime editing are advanced forms of gene-editing technology that offer greater 24. __ than standard CRISPR systems. Unlike conventional CRISPR, which creates breaks in DNA, base editors chemically modify individual DNA letters without cutting. Prime editors can make complex changes including insertions and 25. __. These refined tools reduce unintended mutations and expand the range of 26. __ __ that are possible.


PASSAGE 3 – Ethical Dimensions and Societal Implications of Genetic Interventions

Độ khó: Hard (Band 7.0-9.0)

Thời gian đề xuất: 23-25 phút

The ascendancy of genetic medicine has precipitated a profound ethical discourse that transcends conventional medical ethics and impinges upon fundamental questions about human nature, social justice, and the appropriate boundaries of scientific intervention. As the capability to modify the human germline transitions from theoretical possibility to practical reality, societies worldwide grapple with unprecedented dilemmas that demand multifaceted considerations encompassing philosophical, legal, economic, and cultural dimensions. The ramifications of these technologies extend far beyond individual patients, potentially affecting future generations and the evolutionary trajectory of our species.

The distinction between therapeutic interventions and enhancement technologies constitutes a central axis of ethical debate. While there exists broad consensus regarding the legitimacy of using genetic technologies to treat or prevent serious diseases, the prospect of employing these same tools to enhance normal human traits generates considerable controversy. Enhancement might encompass augmenting cognitive abilities, physical capabilities, longevity, or even aesthetic characteristics. Proponents argue that enhancement represents a natural extension of humanity’s longstanding efforts to improve the human condition through education, nutrition, and medicine. They contend that arbitrary distinctions between therapy and enhancement lack moral coherence, and that procreative liberty encompasses the right to employ genetic technologies to give one’s children advantages. Critics, however, warn that genetic enhancement could exacerbate social inequalities, create pressure for conformity to narrow standards of desirability, and fundamentally alter the concept of what it means to be human.

Justice and equity concerns loom large in discussions about genetic medicine. The exorbitant costs associated with cutting-edge genetic therapies – often exceeding one million dollars per treatment – raise troubling questions about distributive justice. If only affluent individuals can access life-saving or life-enhancing genetic interventions, genetic medicine might crystallize and amplify existing social stratifications, creating a biological underclass unable to afford genetic optimization. This scenario becomes particularly concerning if genetic enhancements affect heritable traits, as socioeconomic advantages could become biologically embedded and transmitted across generations, potentially undermining meritocratic principles and social mobility. Some ethicists advocate for genetic interventions to be treated as public goods, with universal access ensured through collective funding mechanisms. Others question whether societies should prioritize expensive genetic treatments when resources could address more cost-effective public health interventions benefiting larger populations.

The concept of informed consent becomes problematically complex in the context of genetic interventions affecting future individuals who cannot consent to modifications of their genome. When parents make decisions about germline editing that will affect their children and subsequent descendants, they exercise procreative choices with irrevocable multigenerational consequences. Children born with genetically modified germlines had no agency in decisions profoundly affecting their biological inheritance. This raises questions about whether future generations possess rights that constrain contemporary actions, and how to adjudicate conflicts between parental autonomy and children’s interests. Some philosophical frameworks emphasize the right to an open future, suggesting that genetic modifications should not foreclose options or impose predetermined paths upon future individuals. However, determining which modifications respect versus violate this right proves philosophically contentious.

Cultural and religious perspectives significantly influence societal attitudes toward genetic interventions, with substantial heterogeneity across different communities. Some religious traditions view genetic modification as transgressing divine prerogatives or violating the sanctity of natural creation, while others interpret these technologies as extensions of human stewardship and healing obligations. Indigenous communities may hold distinctive perspectives shaped by different conceptualizations of personhood, kinship, and relationship with nature. These diverse worldviews complicate efforts to establish universal ethical frameworks or international regulatory standards. The principle of respect for cultural plurality suggests accommodating diverse approaches, yet genetic technologies that affect the global gene pool may require some degree of international coordination to prevent regulatory arbitrage or unilateral actions with worldwide ramifications.

The prospect of inadvertent genetic discrimination presents another ethical hazard. As genetic information becomes increasingly accessible, concerns arise about its potential misuse by employers, insurers, or other institutions. Although many jurisdictions have enacted genetic information non-discrimination acts, enforcement mechanisms remain imperfect, and subtle forms of discrimination may prove difficult to detect or prosecute. The concept of genetic privacy becomes increasingly nebulous as whole-genome sequencing becomes routine and genetic databases expand. An individual’s genetic information reveals data not only about themselves but also about biological relatives, creating tensions between privacy rights and familial obligations. Furthermore, de-identification of genetic data proves remarkably challenging, as the uniqueness of individual genomes often enables re-identification even from supposedly anonymous datasets.

Regulatory approaches to genetic medicine vary considerably across national jurisdictions, reflecting different risk tolerances, ethical priorities, and governance philosophies. Some countries adopt precautionary stances, imposing stringent restrictions or moratoriums on certain applications, particularly germline editing. Others embrace permissive frameworks that afford scientists considerable latitude within broad ethical guidelines. The United States operates with relatively decentralized regulation, where federal funding restrictions coexist with private sector freedom, creating a complex patchwork of oversight mechanisms. The European Union tends toward risk-averse approaches emphasizing the precautionary principle. Meanwhile, China’s regulatory environment has proven more accommodating of ambitious genetic research, as evidenced by the controversial 2018 creation of gene-edited babies by researcher He Jiankui, which sparked international condemnation and intensified calls for global governance frameworks.

The He Jiankui incident crystallized concerns about the inadequacy of existing regulatory structures and the potential for rogue actors to proceed unilaterally with ethically controversial experiments. He’s unauthorized germline editing of human embryos violated both Chinese regulations and international scientific norms, yet institutional safeguards failed to prevent the research. This episode underscores the need for robust oversight mechanisms, including prospective review of research protocols, transparent reporting requirements, and meaningful penalties for ethical violations. However, designing effective international governance proves formidable given geopolitical tensions, divergent national interests, and the decentralized nature of scientific research. Various stakeholders have proposed international treaties, self-regulatory frameworks within the scientific community, or hybrid models combining governmental oversight with professional self-governance.

Public engagement and democratic deliberation about genetic technologies remain underdeveloped relative to the magnitude of decisions at stake. Technical complexity and specialized knowledge requirements create barriers to meaningful participation by non-expert citizens. Yet decisions about whether and how to employ genetic interventions reflect value judgments that extend beyond technical expertise, encompassing questions about what kind of society we aspire to create and what values should guide technological development. Participatory governance mechanisms such as citizens’ assemblies, consensus conferences, and deliberative polls can help incorporate diverse perspectives and enhance the democratic legitimacy of policy decisions. Effective public engagement requires sustained investment in scientific literacy and creating institutional structures that enable substantive dialogue between experts, policymakers, and publics.

The long-term trajectory of genetic medicine remains inherently uncertain, contingent upon scientific breakthroughs, economic factors, regulatory decisions, and social acceptance. Optimistic projections envision genetic technologies as powerful tools for eliminating suffering, extending healthy lifespans, and enhancing human flourishing. Pessimistic scenarios warn of unforeseen consequences, exacerbated inequalities, and fundamental alterations to human nature that we may later regret. Most likely, the future will involve complex mixtures of benefits and harms, progress and setbacks, requiring ongoing ethical reflection and adaptive governance. The challenge lies in cultivating wisdom to guide these powerful technologies toward beneficial applications while establishing guardrails against misuse and maintaining space for diverse conceptions of human flourishing. As we stand at this pivotal juncture, the decisions we make about genetic medicine will reverberate through generations, shaping not only individual lives but the collective future of humanity.

Hội nghị quốc tế thảo luận về các vấn đề đạo đức trong nghiên cứu chỉnh sửa gen với sự tham gia của các nhà khoa học và chuyên gia y tếHội nghị quốc tế thảo luận về các vấn đề đạo đức trong nghiên cứu chỉnh sửa gen với sự tham gia của các nhà khoa học và chuyên gia y tế

Questions 27-40

Questions 27-31: Multiple Choice

Choose the correct letter, A, B, C, or D.

27. According to the passage, the debate about genetic medicine:

  • A. only concerns medical professionals
  • B. involves multiple dimensions including philosophy and culture
  • C. has been completely resolved
  • D. focuses exclusively on technical issues

28. The distinction between therapy and enhancement is described as:

  • A. universally agreed upon
  • B. irrelevant to ethical discussions
  • C. a central point of ethical debate
  • D. easy to determine in practice

29. Critics of genetic enhancement worry that it might:

  • A. be too expensive for everyone
  • B. worsen social inequalities
  • C. fail to work properly
  • D. take too long to develop

30. The He Jiankui incident demonstrated that:

  • A. germline editing is safe
  • B. current oversight mechanisms have weaknesses
  • C. China has the best regulations
  • D. international cooperation is impossible

31. The passage suggests that public engagement in genetic technology decisions is:

  • A. unnecessary due to technical complexity
  • B. well-developed and effective
  • C. insufficient given the importance of decisions
  • D. impossible to achieve

Questions 32-36: Matching Features

Match each concern (32-36) with the correct ethical dimension (A-G).

Concerns:

32. Only wealthy people can afford expensive treatments

33. Children cannot consent to modifications affecting their genome

34. Genetic information might be misused by employers

35. Different religious traditions have conflicting views

36. Individual genetic data reveals information about relatives

Ethical Dimensions:

  • A. Justice and equity
  • B. Informed consent
  • C. Genetic discrimination
  • D. Cultural and religious perspectives
  • E. Genetic privacy
  • F. Enhancement versus therapy
  • G. Regulatory frameworks

Questions 37-40: Short-answer Questions

Answer the questions below.

Choose NO MORE THAN THREE WORDS from the passage for each answer.

37. What type of genetic modification raises concerns about affecting future generations who cannot consent?

38. What principle do some religious traditions believe genetic modification violates?

39. What did He Jiankui create in 2018 that caused international controversy?

40. What mechanisms are suggested to help include diverse perspectives in policy decisions about genetic technologies?


Answer Keys – Đáp Án

PASSAGE 1: Questions 1-13

  1. B
  2. C
  3. B
  4. A
  5. B
  6. FALSE
  7. TRUE
  8. FALSE
  9. TRUE
  10. defective
  11. simplicity
  12. germline
  13. artificial intelligence

PASSAGE 2: Questions 14-26

  1. YES
  2. NO
  3. NO
  4. YES
  5. NOT GIVEN
  6. ii
  7. iv
  8. v
  9. vi
  10. i
  11. precision
  12. deletions
  13. genetic alterations

PASSAGE 3: Questions 27-40

  1. B
  2. C
  3. B
  4. B
  5. C
  6. A
  7. B
  8. C
  9. D
  10. E
  11. germline editing
  12. natural creation
  13. gene-edited babies
  14. citizens’ assemblies

Giải Thích Đáp Án Chi Tiết

Passage 1 – Giải Thích

Câu 1: B

  • Dạng câu hỏi: Multiple Choice
  • Từ khóa: DNA structure, 1953, important
  • Vị trí trong bài: Đoạn 1, dòng 1-3
  • Giải thích: Bài đọc nói rõ “This groundbreaking discovery… laid the foundation for understanding how genetic information is stored and transmitted”. Đáp án B paraphrase ý này thành “helped scientists understand genetic information storage”.

Câu 2: C

  • Dạng câu hỏi: Multiple Choice
  • Từ khóa: genetic disorders
  • Vị trí trong bài: Đoạn 2, dòng 1-3
  • Giải thích: Câu trong bài viết “These conditions result from abnormalities in an individual’s DNA, which can be inherited from parents or occur spontaneously” tương ứng với đáp án C “may be inherited or occur spontaneously”.

Câu 6: FALSE

  • Dạng câu hỏi: True/False/Not Given
  • Từ khóa: Watson and Crick, 21st century
  • Vị trí trong bài: Đoạn 1, dòng 2
  • Giải thích: Bài viết nói rõ khám phá này diễn ra năm 1953 (thế kỷ 20), không phải thế kỷ 21, nên câu này FALSE.

Câu 10: defective

  • Dạng câu hỏi: Sentence Completion
  • Từ khóa: replace or compensate
  • Vị trí trong bài: Đoạn 3, dòng 2
  • Giải thích: Câu gốc “introducing healthy genes into a patient’s cells to replace or compensate for defective genes” chứa từ cần điền.

Học viên đang tập trung luyện tập bài thi IELTS Reading với tài liệu và máy tính bảng trong không gian học tập hiện đạiHọc viên đang tập trung luyện tập bài thi IELTS Reading với tài liệu và máy tính bảng trong không gian học tập hiện đại

Passage 2 – Giải Thích

Câu 14: YES

  • Dạng câu hỏi: Yes/No/Not Given
  • Từ khóa: understanding molecular mechanisms, essential
  • Vị trí trong bài: Đoạn 1, dòng 1-3
  • Giải thích: Câu đầu tiên của passage khẳng định “Understanding these mechanisms has been crucial for developing targeted therapies that can address the underlying causes of disease”, nghĩa là tác giả đồng ý với quan điểm này.

Câu 15: NO

  • Dạng câu hỏi: Yes/No/Not Given
  • Từ khóa: all genetic diseases, absence of a gene
  • Vị trí trong bài: Đoạn 2, dòng 2-3
  • Giải thích: Bài viết nói “Many diseases result not from the absence of a gene but from its inappropriate expression”, nghĩa là không phải tất cả bệnh di truyền đều do thiếu gen. Điều này mâu thuẫn với câu hỏi.

Câu 19: ii (Paragraph 2)

  • Dạng câu hỏi: Matching Headings
  • Giải thích: Đoạn 2 tập trung vào “gene expression” và các chiến lược để “modulate gene expression” như antisense oligonucleotides, phù hợp với tiêu đề “Modulating gene expression for treatment”.

Câu 24: precision

  • Dạng câu hỏi: Summary Completion
  • Từ khóa: greater
  • Vị trí trong bài: Đoạn 7, dòng 1
  • Giải thích: Câu “Base editing and prime editing represent… offering even greater precision than standard CRISPR systems” chứa từ cần điền.

Passage 3 – Giải Thích

Câu 27: B

  • Dạng câu hỏi: Multiple Choice
  • Từ khóa: debate about genetic medicine
  • Vị trí trong bài: Đoạn 1, dòng 1-5
  • Giải thích: Đoạn mở đầu nói về “profound ethical discourse” với “multifaceted considerations encompassing philosophical, legal, economic, and cultural dimensions”, tương ứng với đáp án B.

Câu 32: A (Justice and equity)

  • Dạng câu hỏi: Matching Features
  • Vị trí trong bài: Đoạn 3
  • Giải thích: Đoạn 3 thảo luận về “Justice and equity concerns” và “exorbitant costs” khiến chỉ người giàu có thể tiếp cận điều trị, phù hợp với concern 32.

Câu 37: germline editing

  • Dạng câu hỏi: Short-answer Questions
  • Từ khóa: future generations, cannot consent
  • Vị trí trong bài: Đoạn 4, dòng 1-2
  • Giải thích: Câu “When parents make decisions about germline editing that will affect their children and subsequent descendants” chứa đáp án.

Từ Vựng Quan Trọng Theo Passage

Passage 1 – Essential Vocabulary

Từ vựng Loại từ Phiên âm Nghĩa tiếng Việt Ví dụ từ bài Collocation
groundbreaking adj /ˈɡraʊndbreɪkɪŋ/ mang tính đột phá This groundbreaking discovery laid the foundation groundbreaking research/discovery
genetic disorder n /dʒəˈnetɪk dɪsˈɔːdə/ rối loạn di truyền Genetic disorders affect millions of people inherited genetic disorder
gene therapy n /dʒiːn ˈθerəpi/ liệu pháp gen The first successful gene therapy trial undergo gene therapy
defective gene n /dɪˈfektɪv dʒiːn/ gen khiếm khuyết To replace defective genes carry a defective gene
milestone n /ˈmaɪlstəʊn/ cột mốc quan trọng Marked another milestone in research significant milestone
clinical trial n /ˈklɪnɪkl ˈtraɪəl/ thử nghiệm lâm sàng Several treatments moved to clinical trials conduct clinical trials
inherited blindness n /ɪnˈherɪtɪd ˈblaɪndnəs/ mù di truyền Treatment for inherited blindness rare form of inherited blindness
blood disorder n /blʌd dɪsˈɔːdə/ rối loạn máu Blood disorders have been amenable treat blood disorders
immune response n /ɪˈmjuːn rɪˈspɒns/ phản ứng miễn dịch Triggered unexpected immune responses adverse immune response
germline editing n /ˈdʒɜːmlaɪn ˈedɪtɪŋ/ chỉnh sửa dòng mầm Concerns about germline editing ban germline editing
human genome n /ˈhjuːmən ˈdʒiːnəʊm/ bộ gen người Understanding of the human genome sequence the human genome

Passage 2 – Essential Vocabulary

Từ vựng Loại từ Phiên âm Nghĩa tiếng Việt Ví dụ từ bài Collocation
molecular basis n /məˈlekjʊlə ˈbeɪsɪs/ cơ sở phân tử The molecular basis of genetic diseases understand molecular basis
gene expression n /dʒiːn ɪkˈspreʃn/ biểu hiện gen Gene expression represents a critical control point regulate gene expression
therapeutic intervention n /ˌθerəˈpjuːtɪk ˌɪntəˈvenʃn/ can thiệp điều trị Critical control point for therapeutic intervention targeted therapeutic intervention
viral vector n /ˈvaɪrəl ˈvektə/ vector virus When viral vectors are introduced engineered viral vector
ex vivo adj/adv /eks ˈviːvəʊ/ ngoài cơ thể sống Ex vivo gene therapy ex vivo modification
CAR T-cell n /kɑː tiː sel/ tế bào T-CAR CAR T-cell therapy exemplifies engineer CAR T-cells
remission rate n /rɪˈmɪʃn reɪt/ tỷ lệ thuyên giảm Achieved remarkable remission rates high remission rate
genetic heterogeneity n /dʒəˈnetɪk ˌhetərəʊdʒəˈniːəti/ tính không đồng nhất di truyền Genetic heterogeneity complicates development account for genetic heterogeneity
precision medicine n /prɪˈsɪʒn ˈmedsn/ y học chính xác Led to the concept of precision medicine develop precision medicine
pharmacogenomics n /ˌfɑːməkəʊdʒɪˈnɒmɪks/ dược lý di truyền Pharmacogenomics helps predict responses apply pharmacogenomics
in vivo adj/adv /ɪn ˈviːvəʊ/ trong cơ thể sống In vivo gene therapy offers advantages in vivo delivery
base editing n /beɪs ˈedɪtɪŋ/ chỉnh sửa base Base editing represents latest evolution perform base editing
off-target effect n /ɒf ˈtɑːɡɪt ɪˈfekt/ tác dụng ngoài mục tiêu Avoiding off-target effects minimize off-target effects
regulatory landscape n /ˈreɡjʊlətri ˈlændskeɪp/ bối cảnh quản lý The regulatory landscape continues to evolve navigate regulatory landscape
manufacturing scalability n /ˌmænjʊˈfækʧərɪŋ ˌskeɪləˈbɪləti/ khả năng mở rộng sản xuất Manufacturing scalability poses hurdles improve manufacturing scalability

Passage 3 – Essential Vocabulary

Từ vựng Loại từ Phiên âm Nghĩa tiếng Việt Ví dụ từ bài Collocation
ascendancy n /əˈsendənsi/ sự lên ngôi, vượt trội The ascendancy of genetic medicine gain ascendancy
precipitate v /prɪˈsɪpɪteɪt/ gây ra, đẩy nhanh Has precipitated a profound ethical discourse precipitate a crisis
germline n /ˈdʒɜːmlaɪn/ dòng mầm Modify the human germline alter the germline
ramification n /ˌræmɪfɪˈkeɪʃn/ hệ quả, phân nhánh The ramifications extend far beyond serious ramifications
therapeutic intervention n /ˌθerəˈpjuːtɪk ˌɪntəˈvenʃn/ can thiệp điều trị Distinction between therapeutic interventions require therapeutic intervention
enhancement technology n /ɪnˈhɑːnsmənt tekˈnɒlədʒi/ công nghệ cải tiến Enhancement technologies generate controversy develop enhancement technologies
procreative liberty n /ˈprəʊkrieɪtɪv ˈlɪbəti/ quyền tự do sinh sản Procreative liberty encompasses the right exercise procreative liberty
distributive justice n /dɪˈstrɪbjʊtɪv ˈdʒʌstɪs/ công lý phân phối Questions about distributive justice principles of distributive justice
exorbitant cost n /ɪɡˈzɔːbɪtənt kɒst/ chi phí cắt cắt The exorbitant costs raise questions face exorbitant costs
biological underclass n /ˌbaɪəˈlɒdʒɪkl ˈʌndəklɑːs/ tầng lớp sinh học thấp Creating a biological underclass form a biological underclass
meritocratic principle n /ˌmerɪtəˈkrætɪk ˈprɪnsəpl/ nguyên tắc trọng tài Undermining meritocratic principles uphold meritocratic principles
informed consent n /ɪnˈfɔːmd kənˈsent/ sự đồng ý được thông báo Concept of informed consent becomes complex obtain informed consent
multigenerational adj /ˌmʌltidʒenəˈreɪʃənl/ đa thế hệ Irrevocable multigenerational consequences multigenerational impact
procreative choice n /ˈprəʊkrieɪtɪv tʃɔɪs/ lựa chọn sinh sản Exercise procreative choices respect procreative choices
regulatory arbitrage n /ˈreɡjʊlətri ˈɑːbɪtrɑːʒ/ kiếm lời từ kẽ hở pháp lý Prevent regulatory arbitrage engage in regulatory arbitrage
genetic discrimination n /dʒəˈnetɪk dɪˌskrɪmɪˈneɪʃn/ phân biệt đối xử di truyền Prospect of inadvertent genetic discrimination prohibit genetic discrimination
precautionary stance n /prɪˈkɔːʃənri stæns/ lập trường thận trọng Adopt precautionary stances take a precautionary stance
rogue actor n /rəʊɡ ˈæktə/ kẻ phá hoại Potential for rogue actors prevent rogue actors
democratic deliberation n /ˌdeməˈkrætɪk dɪˌlɪbəˈreɪʃn/ thảo luận dân chủ Democratic deliberation about technologies promote democratic deliberation

Kết Bài

Chủ đề nghiên cứu di truyền học và khả năng chữa trị bệnh tật đại diện cho một trong những lĩnh vực khoa học tiên tiến nhất hiện nay, và cũng là đề tài xuất hiện thường xuyên trong IELTS Reading Test. Qua bộ đề thi mẫu này, bạn đã được thực hành với 3 passages ở các mức độ khác nhau – từ Easy đến Hard – giúp bạn làm quen với cách IELTS xây dựng đề thi với độ khó tăng dần.

Với 40 câu hỏi đa dạng 7 dạng khác nhau, bạn đã rèn luyện kỹ năng xử lý các dạng bài phổ biến như Multiple Choice, True/False/Not Given, Yes/No/Not Given, Matching Headings, Sentence Completion và Short-answer Questions. Đáp án chi tiết kèm giải thích đã giúp bạn hiểu rõ cách paraphrase, vị trí thông tin và lý do tại sao đáp án đúng/sai.

Phần từ vựng chuyên ngành với hơn 40 từ quan trọng được phân loại theo passage sẽ là tài liệu tham khảo quý giá cho việc học từ vựng theo chủ đề. Hãy thường xuyên ôn tập những từ này cùng với các collocations để nâng cao vốn từ học thuật của mình.

Để đạt kết quả tốt nhất trong IELTS Reading, hãy luyện tập đều đặn với các đề thi mẫu tương tự, quản lý thời gian hiệu quả, và phát triển kỹ năng skimming và scanning. Chúc bạn ôn tập hiệu quả và đạt band điểm cao trong kỳ thi IELTS sắp tới!

Previous Article

IELTS Speaking: Cách Trả Lời "Describe A Time When You Felt Proud Of Your Accomplishments" - Bài Mẫu Band 6-9

Next Article

IELTS Reading: Xu Hướng Làm Việc Từ Xa Toàn Cầu - Đề Thi Mẫu Có Đáp Án Chi Tiết

Write a Comment

Leave a Comment

Email của bạn sẽ không được hiển thị công khai. Các trường bắt buộc được đánh dấu *

Đăng ký nhận thông tin bài mẫu

Để lại địa chỉ email của bạn, chúng tôi sẽ thông báo tới bạn khi có bài mẫu mới được biên tập và xuất bản thành công.
Chúng tôi cam kết không spam email ✨